1
|
Greene G, Zonfa I, Ravasz Regan E. A Boolean network model of hypoxia, mechanosensing and TGF-β signaling captures the role of phenotypic plasticity and mutations in tumor metastasis. PLoS Comput Biol 2025; 21:e1012735. [PMID: 40238833 PMCID: PMC12061430 DOI: 10.1371/journal.pcbi.1012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 05/08/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The tumor microenvironment aids cancer progression by promoting several cancer hallmarks, independent of cancer-related mutations. Biophysical properties of this environment, such as the stiffness of the matrix cells adhere to and local cell density, impact proliferation, apoptosis, and the epithelial to mesenchymal transition (EMT). The latter is a rate-limiting step for invasion and metastasis, enhanced in hypoxic tumor environments but hindered by soft matrices and/or high cell densities. As these influences are often studied in isolation, the crosstalk between hypoxia, biomechanical signals, and the classic EMT driver TGF-β is not well mapped, limiting our ability to predict and anticipate cancer cell behaviors in changing tumor environments. To address this, we built a Boolean regulatory network model that integrates hypoxic signaling with a mechanosensitive model of EMT, which includes the EMT-promoting crosstalk of mitogens and biomechanical signals, cell cycle control, and apoptosis. Our model reproduces the requirement of Hif-1α for proliferation, the anti-proliferative effects of strong Hif-1α stabilization during hypoxia, hypoxic protection from anoikis, and hypoxia-driven mechanosensitive EMT. We offer experimentally testable predictions about the effect of VHL loss on cancer hallmarks, with or without secondary oncogene activation. Taken together, our model serves as a predictive framework to synthesize the signaling responses associated with tumor progression and metastasis in healthy vs. mutant cells. Our single-cell model is a key step towards more extensive regulatory network models that cover damage-response and senescence, integrating most cell-autonomous cancer hallmarks into a single model that can, in turn, control the behavior of in silico cells within a tissue model of epithelial homeostasis and carcinoma.
Collapse
Affiliation(s)
- Grant Greene
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Ian Zonfa
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Erzsébet Ravasz Regan
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| |
Collapse
|
2
|
Piccolo EB, Ge ZD, Filipp ME, Sullivan DP, Thorp EB, Sumagin R. Hypoxia-inducible factor-2α enhances neutrophil survival to promote cardiac injury following myocardial infarction. Am J Physiol Heart Circ Physiol 2024; 327:H1230-H1243. [PMID: 39331023 PMCID: PMC11559636 DOI: 10.1152/ajpheart.00392.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Heart failure is a major cause of mortality following myocardial infarction. Neutrophils are among the first immune cells to accumulate in the infarcted region. Although beneficial functions of neutrophils in heart injury are now appreciated, neutrophils are also well known for their ability to exacerbate inflammation and promote tissue damage. Myocardial infarction induces hypoxia, where hypoxia-inducible factors (HIFs) are activated and play critical roles in cellular functions. In this context, the role of Hif2α in neutrophils during myocardial infarction is unknown. Here, we demonstrate that neutrophil Hif2α deletion markedly attenuates myocardial infarct size, improves cardiac function, reduces neutrophil survival and tissue accumulation, and correlates with increased macrophage engulfment rates. Mechanistic studies revealed that Hif2α promotes neutrophil survival through binding to hypoxia response element (HRE) in the promoter region of Birc2 to regulate expression of the prosurvival factor, cellular inhibitor of apoptosis protein-1 (cIAP1). Inhibition of cIAP1 in neutrophils using the pharmacological agent, Birinapant resulted in increased cell death, establishing a critical role of cIAP1 downstream of Hif2α in neutrophil survival. Taken together, our data demonstrate a protective effect of Hif2α deletion in neutrophils on cardiac injury outcomes through modulation of neutrophil cell survival.NEW & NOTEWORTHY Hif2α in neutrophils increases infarct size, cardiac dysfunction, and ventricular scar after myocardial infarction. Hif2α in neutrophils supports neutrophil survival via cIAP-1 signaling and delays macrophage engulfment.
Collapse
Affiliation(s)
- Enzo B Piccolo
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | | | - Mallory E Filipp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - David P Sullivan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| |
Collapse
|
3
|
Zhang W, Yang F, Yan Q, Li J, Zhang X, Jiang Y, Dai J. Hypoxia inducible factor-1α related mechanism and TCM intervention in process of early fracture healing. CHINESE HERBAL MEDICINES 2024; 16:56-69. [PMID: 38375046 PMCID: PMC10874770 DOI: 10.1016/j.chmed.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 02/21/2024] Open
Abstract
As a common clinical disease, fracture is often accompanied by pain, swelling, bleeding as well as other symptoms and has a high disability rate, even threatening life, seriously endangering patients' physical and psychological health and quality of life. Medical practitioners take many strategies for the treatment of fracture healing, including Traditional Chinese Medicine (TCM). In the early stage of fracture healing, the local fracture is often in a state of hypoxia, accompanied by the expression of hypoxia inducible factor-1α (HIF-1α), which is beneficial to wound healing. Through literature mining, we thought that hypoxia, HIF-1α and downstream factors affected the mechanism of fracture healing, as well as dominated this process. Therefore, we reviewed the local characteristics and related signaling pathways involved in the fracture healing process and summarized the intervention of TCM on these mechanisms, in order to inspirit the new strategy for fracture healing, as well as elaborate on the possible principles of TCM in treating fractures based on the HIF molecular mechanism.
Collapse
Affiliation(s)
- Wenxian Zhang
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Fusen Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Qikai Yan
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an 710021, China
| | - Jiahui Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiaogang Zhang
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Yiwei Jiang
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Jianye Dai
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
4
|
Park SG, Ji MJ, Ham IH, Shin YH, Lee SM, Lee CH, Kim E, Hur H, Park HM, Kim JY. Secretome analysis reveals reduced expression of COL4A2 in hypoxic cancer-associated fibroblasts with a tumor-promoting function in gastric cancer. J Cancer Res Clin Oncol 2023; 149:4477-4487. [PMID: 36125535 DOI: 10.1007/s00432-022-04361-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are major components of the tumor microenvironment (TME). Hypoxic TME is known to promote tumor progression. However, how a hypoxic condition regulates CAFs remains elusive. METHODS To investigate the underlying mechanism involved in the regulation of gastric cancer (GC) progression by hypoxic CAFs, we performed secretome profiling. Normoxic or hypoxic CAFs conditioned media (CM) were filter-concentrated and in-gel trypsin digested. Resulting peptides were analyzed with LC-MS/MS. RESULTS We observed that CM derived from hypoxic CAFs could promote migration of a panel of GC cell lines (AGS, SNU668, SNU638). Mass spectrometry analysis of hypoxic or normoxic CAFs CM identified 1595 proteins, of which 19 proteins (10 upregulated and 9 downregulated) were differentially expressed in the hypoxic secretome. We focused on COL4A2, whose expression was significantly decreased in hypoxic CAFs in HIF-1α-independent manner. Silencing of COL4A2 expression in normoxic CAFs phenocopied the effect of hypoxic CAFs in promoting GC cell migration. CONCLUSIONS The reduced expression of COL4A2 in a hypoxic environment might be associated with the tumor-promoting role of hypoxic CAFs in GC.
Collapse
Affiliation(s)
- Seo-Gyu Park
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Mi-Jung Ji
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Inflammaging Translational Research Cancer, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Yoon-Hee Shin
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Su-Min Lee
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Chang Hoon Lee
- Therapeutics and Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
- R and D center, SCBIO Co. Ltd, Daejeon, 34050, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Inflammaging Translational Research Cancer, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Department of Biomedical Science, Graduated School of Ajou University, Suwon, 16499, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
5
|
High oxygen-modified packaging (HiOx-MAP) mediates HIF-1α regulation of tenderness changes during postmortem aging of yak meat. Food Chem X 2023; 17:100573. [PMID: 36845525 PMCID: PMC9945635 DOI: 10.1016/j.fochx.2023.100573] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
In the present study, we studied the effect of high oxygen-modified packaging (HiOx-MAP) on yak meat tenderness and the underlying mechanism. HiOx-MAP significantly increased the myofibril fragmentation index (MFI) of yak meat. In addition, western blotting showed that the expression of hypoxia-inducible factor (HIF-1α) and ryanodine receptors (RyR) in the HiOx-MAP group was reduced. HiOx-MAP increased the activity of sarcoplasmic reticulum calcium-ATPase (SERCA). The energy disperse spectroscopy (EDS) mapping showed gradually reduced calcium distribution in the treated endoplasmic reticulum. Furthermore, HiOx-MAP treatment increased the caspase-3 activity and the apoptosis rate. The activity of calmodulin protein (CaMKKβ) and AMP-activated protein kinase (AMPK) was down-regulated leading to apoptosis. These results indicated that HiOx-MAP promoted apoptosis during postmortem aging to improve the tenderization of meat.
Collapse
|
6
|
Ucaryilmaz Metin C, Ozcan G. The HIF-1α as a Potent Inducer of the Hallmarks in Gastric Cancer. Cancers (Basel) 2022; 14:2711. [PMID: 35681691 PMCID: PMC9179860 DOI: 10.3390/cancers14112711] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxia is the principal architect of the topographic heterogeneity in tumors. Hypoxia-inducible factor-1α (HIF-1α) reinforces all hallmarks of cancer and donates cancer cells with more aggressive characteristics at hypoxic niches. HIF-1α potently induces sustained growth factor signaling, angiogenesis, epithelial-mesenchymal transition, and replicative immortality. Hypoxia leads to the selection of cancer cells that evade growth suppressors or apoptotic triggers and deregulates cellular energetics. HIF-1α is also associated with genetic instability, tumor-promoting inflammation, and escape from immunity. Therefore, HIF-1α may be an important therapeutic target in cancer. Despite that, the drug market lacks safe and efficacious anti-HIF-1α molecules, raising the quest for fully unveiling the complex interactome of HIF-1α in cancer to discover more effective strategies. The knowledge gap is even wider in gastric cancer, where the number of studies on hypoxia is relatively low compared to other well-dissected cancers. A comprehensive review of the molecular mechanisms by which HIF-1α induces gastric cancer hallmarks could provide a broad perspective to the investigators and reveal missing links to explore in future studies. Thus, here we review the impact of HIF-1α on the cancer hallmarks with a specific focus on gastric cancer.
Collapse
Affiliation(s)
| | - Gulnihal Ozcan
- Department of Medical Pharmacology, School of Medicine, Koç University, 34450 Istanbul, Turkey
| |
Collapse
|
7
|
Kimura T, Sakai M, Gojo N, Watanabe M, Uzawa N, Sakai T. The HIF-1α pathway plays a critical role in salivary gland development in ex vivo organ cultures. FEBS Open Bio 2021; 12:460-469. [PMID: 34904400 PMCID: PMC8804608 DOI: 10.1002/2211-5463.13351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/25/2021] [Accepted: 12/11/2021] [Indexed: 12/15/2022] Open
Abstract
The transcription factor, hypoxia‐inducible factor‐1α (HIF‐1α), has previously been shown to upregulate the expression of hypoxia‐related genes, including erythropoietin (EPO). However, the role of hypoxia‐inducible factor‐1α in morphogenesis during salivary gland development is unclear. We investigated the function of HIF‐1α in submandibular gland (SMG) organ cultures obtained from embryonic day 13.5 embryos from ICR female mice. Expression of HIF‐1α, glucose transporter 1, and vascular endothelial growth factor was induced under hypoxia (5% O2). We further showed that BAY 87‐2243‐mediated inhibition of HIF‐1α suppressed salivary gland development. Under severe hypoxia (1% O2), HIF‐1α did not promote salivary gland development; this was due to suppression of cell proliferation and inhibition of the cell cycle and not because of autophagy and apoptosis. Additionally, using the inhibitor U0126, we verified that the ERK1/2 pathway is upstream of HIF‐1α. Overall, we found that the HIF‐1α signaling pathway plays a critical role in salivary gland development in ex vivo SMG organ cultures.
Collapse
Affiliation(s)
- Tomomasa Kimura
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan.,Department of Oral and Maxillofacial Surgery 2, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Manabu Sakai
- Department of Clinical Laboratory, Osaka University Dental Hospital, Suita, Japan.,Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Nao Gojo
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Mikio Watanabe
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Narikazu Uzawa
- Department of Oral and Maxillofacial Surgery 2, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Takayoshi Sakai
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan
| |
Collapse
|
8
|
Nwahara N, Abrahams G, Prinsloo E, Nyokong T. Folic acid-modified phthalocyanine-nanozyme loaded liposomes for targeted photodynamic therapy. Photodiagnosis Photodyn Ther 2021; 36:102527. [PMID: 34517120 DOI: 10.1016/j.pdpdt.2021.102527] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/29/2021] [Accepted: 09/07/2021] [Indexed: 01/25/2023]
Abstract
The hypoxic tumour microenvironment and poor spatiotemporal localization of photosensitizers are two significant obstacles that limit practical applications of photodynamic therapy. In response, a biocompatible, light-activatable liposome integrated with both a zinc phthalocyanine photodynamic component and Pt nanoparticles-decorated with MnO2 catalase-mimicking component are engineered. This multifunctional system was rationally designed using unsaturated phospholipids to achieve on-demand drug release following light irradiation. Specificity was achieved by folic acid functionalization resulting in folate-modified liposomes (FTLiposomes). We demonstrated its specific uptake by fluorescence imaging using folate receptor (FR) overexpressing HeLa and MCF-7 cells as in vitro models. This multifunctional liposome exhibits superior hypoxic anti-tumour effects and holds the potential to reduce side effects associated with untargeted therapy. Fluorescence of the constituent ZnPc and folate-receptor targeting could enable tracking and permit spatiotemporal regulation for improved cancer treatment.
Collapse
Affiliation(s)
| | - Garth Abrahams
- Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| | | | | |
Collapse
|
9
|
Coates BS, Deleury E, Gassmann AJ, Hibbard BE, Meinke LJ, Miller NJ, Petzold-Maxwell J, French BW, Sappington TW, Siegfried BD, Guillemaud T. Up-regulation of apoptotic- and cell survival-related gene pathways following exposures of western corn rootworm to B. thuringiensis crystalline pesticidal proteins in transgenic maize roots. BMC Genomics 2021; 22:639. [PMID: 34479486 PMCID: PMC8418000 DOI: 10.1186/s12864-021-07932-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/04/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Resistance of pest insect species to insecticides, including B. thuringiensis (Bt) pesticidal proteins expressed by transgenic plants, is a threat to global food security. Despite the western corn rootworm, Diabrotica virgifera virgifera, being a major pest of maize and having populations showing increasing levels of resistance to hybrids expressing Bt pesticidal proteins, the cell mechanisms leading to mortality are not fully understood. RESULTS Twenty unique RNA-seq libraries from the Bt susceptible D. v. virgifera inbred line Ped12, representing all growth stages and a range of different adult and larval exposures, were assembled into a reference transcriptome. Ten-day exposures of Ped12 larvae to transgenic Bt Cry3Bb1 and Gpp34/Tpp35Ab1 maize roots showed significant differential expression of 1055 and 1374 transcripts, respectively, compared to cohorts on non-Bt maize. Among these, 696 were differentially expressed in both Cry3Bb1 and Gpp34/Tpp35Ab1 maize exposures. Differentially-expressed transcripts encoded protein domains putatively involved in detoxification, metabolism, binding, and transport, were, in part, shared among transcripts that changed significantly following exposures to the entomopathogens Heterorhabditis bacteriophora and Metarhizium anisopliae. Differentially expressed transcripts in common between Bt and entomopathogen treatments encode proteins in general stress response pathways, including putative Bt binding receptors from the ATP binding cassette transporter superfamily. Putative caspases, pro- and anti-apoptotic factors, as well as endoplasmic reticulum (ER) stress-response factors were identified among transcripts uniquely up-regulated following exposure to either Bt protein. CONCLUSIONS Our study suggests that the up-regulation of genes involved in ER stress management and apoptotic progression may be important in determining cell fate following exposure of susceptible D. v. virgifera larvae to Bt maize roots. This study provides novel insights into insect response to Bt intoxication, and a possible framework for future investigations of resistance mechanisms.
Collapse
Affiliation(s)
- Brad S Coates
- USDA-ARS, Corn Insects & Crop Genetics Research Unit, 103 Genetics Laboratory, Iowa State University, Ames, IA, 50011, USA.
| | | | | | | | - Lance J Meinke
- Department of Entomology, University of Nebraska, Lincoln, NE, USA
| | | | | | - B Wade French
- USDA-ARS, North Central Agricultural Research Laboratory, Brookings, SD, USA
| | - Thomas W Sappington
- USDA-ARS, Corn Insects & Crop Genetics Research Unit, 103 Genetics Laboratory, Iowa State University, Ames, IA, 50011, USA
| | | | | |
Collapse
|
10
|
Prikhodko VA, Selizarova NO, Okovityi SV. [Molecular mechanisms for hypoxia development and adaptation to it. Part I]. Arkh Patol 2021; 83:52-61. [PMID: 33822555 DOI: 10.17116/patol20218302152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hypoxia is a typical pathological process characterized by the occurrence of oxygen deficiency in tissues and cells and accompanied by the development of immediate and delayed compensatory and adaptive reactions. Reprogramming of the mitochondrial electron transport chain (ETC) function is one the most essential regulatory mechanisms that allow for immediate adaptation to hypoxia. Succinic acid, or succinate, is involved in this process not only as one of the intermediates of the tricarboxylic acid (TAC) cycle, but also as a signaling molecule. In this connection, the purpose of this review was to systematize the available data on the molecular mechanisms for the development of hypoxia and its adaptation at the ETC/TAC coupling site, as well as on the role of succinic acid in these processes.
Collapse
Affiliation(s)
- V A Prikhodko
- Saint Petersburg State Chemical and Pharmaceutical University of the Ministry of Health of the Russian Federation, Saint Petersburg, Russia
| | - N O Selizarova
- Saint Petersburg State Chemical and Pharmaceutical University of the Ministry of Health of the Russian Federation, Saint Petersburg, Russia
| | - S V Okovityi
- Saint Petersburg State Chemical and Pharmaceutical University of the Ministry of Health of the Russian Federation, Saint Petersburg, Russia
| |
Collapse
|
11
|
Gu X, Meng H, Wang J, Wang R, Cao M, Liu S, Chen H, Xu Y. Hypoxia contributes to galectin-3 expression in renal carcinoma cells. Eur J Pharmacol 2020; 890:173637. [PMID: 33065093 DOI: 10.1016/j.ejphar.2020.173637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023]
Abstract
Galectin-3 is supposed as a prognostic factor and therapeutic target for many cancers. In a previous study, we have reported that galectin-3 was related to the development of renal cell cancer and served a therapeutic target for renal cell carcinoma (RCC). However, the mechanisms underlying the regulation of galectin-3 in RCC are still not known. In this study, we detected the expression of galectin-3 and hypoxia-inducible factor 1 (HIF-1) α in RCC using immunohistochemistry, and then conducted in vitro experiments to verify the regulation of galectin-3 by hypoxia in RCC. Our results showed that the expression of galectin-3 and HIF-1α were remarkably high in RCC tissues compared with those in the paracancerous tissues. Interestingly, hypoxia significantly promoted cytoplasmic and nuclear HIF-1α and galectin-3 expression in renal carcinoma cell lines, but not in renal tubular epithelial cell (HK-2). Renal carcinoma cell line (Caki-1), but not HK-2 showed significant increase of luciferase reporter activity of galectin-3 encoding the fragment from the site of -845 to +50 upon hypoxic insult. Moreover, HIF-1α overexpression vector promoted, while HIF-1α silencing vector reduced luciferase reporter activity of galectin-3 in Caki-1 and HK-2 cells in both normal and hypoxia conditions. A direct interaction of HIF-1α with Gal-3 promoter was also verified by electrophoretic mobility shift assay and chromatin immunoprecipitation. Together, our data indicated that hypoxia was critical for galectin-3 expression in RCC in a HIF-1α-dependent manner.
Collapse
Affiliation(s)
- Xin Gu
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jia Wang
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Ruitao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Muyang Cao
- Department of Urology Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Siyu Liu
- Department of Urology Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Hui Chen
- Department of Urology Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Yangyang Xu
- Department of Urology Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
12
|
Geng X, Xu X, Fang Y, Zhao S, Hu J, Xu J, Jia P, Ding X, Teng J. Effect of long non-coding RNA growth arrest-specific 5 on apoptosis in renal ischaemia/reperfusion injury. Nephrology (Carlton) 2019; 24:405-413. [PMID: 30129267 DOI: 10.1111/nep.13476] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
AIM Long non-coding RNA (lncRNAs) have been shown to play a critical role in a variety of pathophysiological processes, such as cell proliferation, apoptosis and migration. However, there were few studies addressing the function of lncRNAs in renal ischaemia/reperfusion (I/R) injury. Apoptosis is an important pathogenesis during I/R injury. Here, we identified the effect of hypoxia-responsive lncRNA growth arrest-specific 5 (GAS5) on apoptosis in renal I/R injury. METHODS Ischaemia/reperfusion injury in mice or hypoxia/re-oxygenation (H/R) in human proximal renal tubular epithelial cells (HK-2) was practiced to induce apoptosis. The kidneys and blood were collected at 24 h after reperfusion. The GAS5 messenger RNA (mRNA) expression and apoptosis-related gene mRNA and protein levels, including p53, cellular inhibitor of apoptosis protein 2 (cIAP2) and thrombospondin-1 (TSP-1), were analysed. GAS5 small-interfering RNA was transfected with H/R induced cells. Over-expression of GAS5 was performed by plasmid transfection. RESULTS Apoptotic cells significantly increased in I/R-injured kidneys. GAS5 could be up-regulated in kidneys at 24 h after reperfusion and 3 h after re-oxygenation, combined with increased expression of its downstream apoptosis-related proteins p53 and cIAP2. GAS5 small-interfering RNA treatment down-regulated the mRNA and protein levels of p53 and TSP-1, and attenuated apoptosis induced by H/R in HK-2 cells. Conversely, over-expression of GAS5 up-regulated the mRNA and protein levels of p53 and TSP-1, and promoted apoptosis in HK-2 cells. CONCLUSION Long non-coding RNA GAS5 induced by I/R injury could promote apoptosis in kidney. TSP-1 might be one of the downstream effectors of GAS5, which will be explored in the future.
Collapse
Affiliation(s)
- Xuemei Geng
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jiachang Hu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jiarui Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Ping Jia
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| |
Collapse
|
13
|
Soubéran A, Cappaï J, Chocry M, Nuccio C, Raujol J, Colin C, Lafitte D, Kovacic H, Quillien V, Baeza-Kallee N, Rougon G, Figarella-Branger D, Tchoghandjian A. Inhibitor of Apoptosis Proteins Determines Glioblastoma Stem-Like Cell Fate in an Oxygen-Dependent Manner. Stem Cells 2019; 37:731-742. [PMID: 30920104 DOI: 10.1002/stem.2997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
In glioblastomas, apoptosis inhibitor proteins (IAPs) are involved in apoptotic and nonapoptotic processes. We previously showed that IAP inhibition induced a loss of stemness and glioblastoma stem cells differentiation by activating nuclear factor-κB under normoxic conditions. Hypoxia has been shown to modulate drug efficacy. Here, we investigated how IAPs participate in glioblastoma stem-like cell maintenance and fate under hypoxia. We showed that in a hypoxic environment, IAPs inhibition by GDC-0152, a small-molecule IAPs inhibitor, triggered stem-like cell apoptosis and decreased proliferation in four human glioblastoma cell lines. We set up a three-dimensional glioblastoma spheroid model in which time-of-flight secondary ion mass spectrometry analyses revealed a decrease in oxygen levels between the periphery and core. We observed low proliferative and apoptotic cells located close to the hypoxic core of the spheres and glial fibrillary acidic protein+ cells at their periphery. These oxygen-dependent GDC-0152 antitumoral effects have been confirmed on human glioblastoma explants. Notably, serine-threonine kinase activation analysis revealed that under hypoxic conditions, IAP inhibition activated ataxia telangiectasia and Rad3-related protein signaling. Our findings provide new insights into the dual mechanism of action of IAP inhibitors that depends on oxygen level and are relevant to their therapeutic application in tumors. Stem Cells 2019;37:731-742.
Collapse
Affiliation(s)
- Aurélie Soubéran
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Jessica Cappaï
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Mathieu Chocry
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | | | | | - Carole Colin
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Daniel Lafitte
- Aix-Marseille University, INSERM UMR MD1, Marseille, France
| | - Hervé Kovacic
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Véronique Quillien
- Rennes 1 University, "Chemistry, Oncogenesis, Stress, Signaling", INSERM U1242, Rennes, France.,Centre de lutte contre le cancer Eugène Marquis, Rennes F-35042, France
| | - Nathalie Baeza-Kallee
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Geneviève Rougon
- Aix-Marseille University, Institut de Neurosciences Timone, CNRS 7289, Marseille, France
| | - Dominique Figarella-Branger
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,AP-HM, Timone Hospital, Department of Anatomopathology, Marseille, France
| | - Aurélie Tchoghandjian
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| |
Collapse
|
14
|
STAT3 but Not HIF-1α Is Important in Mediating Hypoxia-Induced Chemoresistance in MDA-MB-231, a Triple Negative Breast Cancer Cell Line. Cancers (Basel) 2017; 9:cancers9100137. [PMID: 29036915 PMCID: PMC5664076 DOI: 10.3390/cancers9100137] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/25/2022] Open
Abstract
Hypoxia-induced chemoresistance (HICR) is a well-recognized phenomenon, and in many experimental models, hypoxia inducible factor-1α (HIF-1α) is believed to be a key player. We aimed to better understand the mechanism underlying HICR in a triple negative breast cancer cell line, MDA-MB-231, with a focus on the role of HIF-1α. In this context, the effect of hypoxia on the sensitivity of MDA-MB-231 cells to cisplatin and their stem-like features was evaluated and the role of HIF-1α in both phenomena was assessed. Our results showed that hypoxia significantly increased MDA-MB-231 resistance to cisplatin. Correlating with this, intracellular uptake of cisplatin was significantly reduced under hypoxia. Furthermore, the stem-like features of MDA-MB-231 cells increased as evidenced by the significant increases in the expression of ATP-binding cassette (ABC) drug transporters, the proportion of CD44+/CD24− cells, clonogenic survival and cisplatin chemoresistance. Under hypoxia, both the protein level and DNA binding of HIF-1α was dramatically increased. Surprisingly, siRNA knockdown of HIF-1α did not result in an appreciable change to HICR. Instead, signal transducer and activator of transcription 3 (STAT3) activation was found to be important. STAT3 activation may confer HICR by upregulating ABC transporters, particularly ABCC2 and ABCC6. This study has demonstrated that, in MDA-MB-231 cells, STAT3 rather than HIF-1α is important in mediating HICR to cisplatin.
Collapse
|
15
|
Targeting Tumor Adaption to Chronic Hypoxia: Implications for Drug Resistance, and How It Can Be Overcome. Int J Mol Sci 2017; 18:ijms18091854. [PMID: 28841148 PMCID: PMC5618503 DOI: 10.3390/ijms18091854] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022] Open
Abstract
The rapid and uncontrolled proliferation of tumors limits the availability of oxygen and nutrients supplied from the tumor vasculature, thus exposing them to low oxygen environments. Thus, diminished oxygen availability, or hypoxia, is the most common microenvironment feature of nearly all solid tumors. All living cells have the ability to sense changes in oxygen tension and adapt to this stress to preserve survival. Likewise, cancer cells adapt to chronic hypoxic stress via several mechanisms, including promotion of angiogenic factor production, metabolic shift to consume less oxygen, and reduction of apoptotic potential. Adaptation of tumor cells to hypoxia is believed to be the main driver for selection of more invasive and therapy-resistant cancer phenotypes. In this review, we discuss molecular mechanisms by which tumor cells adapt to hypoxia, with a specific focus on hypoxia-inducible factor (HIF) transcription factor. We further discuss the current understandings on hypoxia-mediated drug resistance and strategies to overcome it.
Collapse
|
16
|
Tak E, Jun DY, Kim SH, Park GC, Lee J, Hwang S, Song GW, Lee SG. Upregulation of P2Y2 nucleotide receptor in human hepatocellular carcinoma cells. J Int Med Res 2016; 44:1234-1247. [PMID: 27807254 PMCID: PMC5536769 DOI: 10.1177/0300060516662135] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective To examine if hypoxia inducible factor-1α (HIF-1α) can induce the upregulation of the purinergic receptor P2Y2 (P2Y2) and thereby promote the viability of human hepatocellular carcinoma (HCC) cells under hypoxic conditions. Methods Archival HCC tumour specimens and corresponding non-cancerous tissues were examined immunohistochemically for P2Y2 protein. A series of in vitro experiments were undertaken using HCC cell lines to determine the effect of hypoxia on HIF-1α and P2Y2 levels, the effect of HIF-1α upregulation on P2Y2 levels, and the effect of P2Y2 upregulation on cell viability under hypoxic conditions. Results Human HCC specimens were positive for P2Y2. Hypoxia and upregulated HIF-1α both upregulated the P2Y2 levels in HCC cell lines. P2Y2 upregulation using plasmid transfection resulted in enhanced cell viability under hypoxia. Treatment of HepG2 cells with the selective P2Y2 antagonist MRS2312 downregulated P2Y2 and reduced cell viability in five HCC cell lines. P2Y2 knockdown reduced HepG2 cell viability under hypoxia. Conclusions These present results suggest that HCC cells upregulate P2Y2 levels during hypoxia, which in turn promotes their growth. P2Y2 could be a potential therapeutic target for treating HCC.
Collapse
Affiliation(s)
- Eunyoung Tak
- 1 Asan Institute for Life Sciences, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae Young Jun
- 1 Asan Institute for Life Sciences, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seok-Hwan Kim
- 2 Department of Surgery, Division of Liver Transplantation and Hepatobiliary Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gil-Chun Park
- 2 Department of Surgery, Division of Liver Transplantation and Hepatobiliary Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jooyoung Lee
- 1 Asan Institute for Life Sciences, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Shin Hwang
- 2 Department of Surgery, Division of Liver Transplantation and Hepatobiliary Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gi-Won Song
- 2 Department of Surgery, Division of Liver Transplantation and Hepatobiliary Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Gyu Lee
- 2 Department of Surgery, Division of Liver Transplantation and Hepatobiliary Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Alonso J, Galán M, Martí-Pàmies I, Romero JM, Camacho M, Rodríguez C, Martínez-González J. NOR-1/NR4A3 regulates the cellular inhibitor of apoptosis 2 (cIAP2) in vascular cells: role in the survival response to hypoxic stress. Sci Rep 2016; 6:34056. [PMID: 27654514 PMCID: PMC5032021 DOI: 10.1038/srep34056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/07/2016] [Indexed: 12/16/2022] Open
Abstract
Vascular cell survival is compromised under pathological conditions such as abdominal aortic aneurysm (AAA). We have previously shown that the nuclear receptor NOR-1 is involved in the survival response of vascular cells to hypoxia. Here, we identify the anti-apoptotic protein cIAP2 as a downstream effector of NOR-1. NOR-1 and cIAP2 were up-regulated in human AAA samples, colocalizing in vascular smooth muscle cells (VSMC). While NOR-1 silencing reduced cIAP2 expression in vascular cells, lentiviral over-expression of this receptor increased cIAP2 mRNA and protein levels. The transcriptional regulation of the human cIAP2 promoter was analyzed in cells over-expressing NOR-1 by luciferase reporter assays, electrophoretic mobility shift analysis and chromatin immunoprecipitation, identifying a NGFI-B site (NBRE-358/-351) essential for NOR-1 responsiveness. NOR-1 and cIAP2 were up-regulated by hypoxia and by a hypoxia mimetic showing a similar time-dependent pattern. Deletion and site-directed mutagenesis studies show that NOR-1 mediates the hypoxia-induced cIAP2 expression. While NOR-1 over-expression up-regulated cIAP2 and limited VSMC apoptosis induced by hypoxic stress, cIAP2 silencing partially prevented this NOR-1 pro-survival effect. These results indicate that cIAP2 is a target of NOR-1, and suggest that this anti-apoptotic protein is involved in the survival response to hypoxic stress mediated by NOR-1 in vascular cells.
Collapse
Affiliation(s)
- Judith Alonso
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - María Galán
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.,Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Ingrid Martí-Pàmies
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - José María Romero
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Mercedes Camacho
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - José Martínez-González
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| |
Collapse
|
18
|
Petit C, Gouel F, Dubus I, Heuclin C, Roget K, Vannier JP. Hypoxia promotes chemoresistance in acute lymphoblastic leukemia cell lines by modulating death signaling pathways. BMC Cancer 2016; 16:746. [PMID: 27658583 PMCID: PMC5034444 DOI: 10.1186/s12885-016-2776-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 08/26/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Several studies show that bone marrow (BM) microenvironment and hypoxia condition can promote the survival of leukemic cells and induce resistance to anti-leukemic drugs. However, the molecular mechanism for chemoresistance by hypoxia is not fully understood. METHODS In the present study, we investigated the effect of hypoxia on resistance to two therapies, methotrexate (MTX) and prednisolone (PRD), in two cell models for acute lymphoblastic leukemia (ALL). To look for an implication of hypoxia in chemoresistance, cell viability, total cell density and cell proliferation were analyzed. Survival and death signaling pathways were also screened by "reverse phase protein array" (RPPA) and western blotting experiments conducted on selected proteins to confirm the results. RESULTS We found that hypoxia promotes chemoresistance in both ALL cell lines. The induction of drug-resistance by hypoxia was not associated with an increase in total cell density nor an increase in cell proliferation. Using RPPA, we show that chemoresistance induced by hypoxia was mediated through an alteration of cell death signaling pathways. This protective effect of hypoxia seems to occur via a decrease in pro-apoptotic proteins and an increase in anti-apoptotic proteins. The results were confirmed by immunoblotting. Indeed, hypoxia is able to modulate the expression of anti-apoptotic proteins independently of chemotherapy while a pro-apoptotic signal induced by a chemotherapy is not modulated by hypoxia. CONCLUSIONS Hypoxia is a factor in leukemia cell resistance and for two conventional chemotherapies modulates cell death signaling pathways without affecting total cell density or cell proliferation.
Collapse
Affiliation(s)
- C. Petit
- MERCI EA3829, Université de Rouen, Faculté de Médecine-Pharmacie, 22 Boulevard Gambetta, 76183 Rouen, France
- BioSIMS Technologies, 75 Route de Lyons la forêt, Seine BioPolis, 76000 Rouen, France
| | - F. Gouel
- MERCI EA3829, Université de Rouen, Faculté de Médecine-Pharmacie, 22 Boulevard Gambetta, 76183 Rouen, France
| | - I. Dubus
- MERCI EA3829, Université de Rouen, Faculté de Médecine-Pharmacie, 22 Boulevard Gambetta, 76183 Rouen, France
| | - C. Heuclin
- BioSIMS Technologies, 75 Route de Lyons la forêt, Seine BioPolis, 76000 Rouen, France
| | - K. Roget
- Enterome, 94/96 avenue Ledru-Rollin, 75011 Paris, France
| | - J. P. Vannier
- MERCI EA3829, Université de Rouen, Faculté de Médecine-Pharmacie, 22 Boulevard Gambetta, 76183 Rouen, France
| |
Collapse
|
19
|
Matsuura K, Canfield K, Feng W, Kurokawa M. Metabolic Regulation of Apoptosis in Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 327:43-87. [PMID: 27692180 DOI: 10.1016/bs.ircmb.2016.06.006] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Apoptosis is a cellular suicide program that plays a critical role in development and human diseases, including cancer. Cancer cells evade apoptosis, thereby enabling excessive proliferation, survival under hypoxic conditions, and acquired resistance to therapeutic agents. Among various mechanisms that contribute to the evasion of apoptosis in cancer, metabolism is emerging as one of the key factors. Cellular metabolites can regulate functions of pro- and antiapoptotic proteins. In turn, p53, a regulator of apoptosis, also controls metabolism by limiting glycolysis and facilitating mitochondrial respiration. Consequently, with dysregulated metabolism and p53 inactivation, cancer cells are well-equipped to disable the apoptotic machinery. In this article, we review how cellular apoptosis is regulated and how metabolism can influence the signaling pathways leading to apoptosis, especially focusing on how glucose and lipid metabolism are altered in cancer cells and how these alterations can impact the apoptotic pathways.
Collapse
Affiliation(s)
- K Matsuura
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| | - K Canfield
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - W Feng
- Norris Cotton Cancer Center, Lebanon, NH, United States
| | - M Kurokawa
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; Norris Cotton Cancer Center, Lebanon, NH, United States.
| |
Collapse
|
20
|
Gerber VEM, Wijenayake S, Storey KB. Anti-apoptotic response during anoxia and recovery in a freeze-tolerant wood frog (Rana sylvatica). PeerJ 2016; 4:e1834. [PMID: 27042393 PMCID: PMC4811176 DOI: 10.7717/peerj.1834] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/01/2016] [Indexed: 11/29/2022] Open
Abstract
The common wood frog, Rana sylvatica, utilizes freeze tolerance as a means of winter survival. Concealed beneath a layer of leaf litter and blanketed by snow, these frogs withstand subzero temperatures by allowing approximately 65–70% of total body water to freeze. Freezing is generally considered to be an ischemic event in which the blood oxygen supply is impeded and may lead to low levels of ATP production and exposure to oxidative stress. Therefore, it is as important to selectively upregulate cytoprotective mechanisms such as the heat shock protein (HSP) response and expression of antioxidants as it is to shut down majority of ATP consuming processes in the cell. The objective of this study was to investigate another probable cytoprotective mechanism, anti-apoptosis during oxygen deprivation and recovery in the anoxia tolerant wood frog. In particular, relative protein expression levels of two important apoptotic regulator proteins, Bax and p-p53 (S46), and five anti-apoptotic/pro-survival proteins, Bcl-2, p-Bcl-2 (S70), Bcl-xL, x-IAP, and c-IAP in response to normoxic, 24 Hr anoxic exposure, and 4 Hr recovery stages were assessed in the liver and skeletal muscle using western immunoblotting. The results suggest a tissue-specific regulation of the anti-apoptotic pathway in the wood frog, where both liver and skeletal muscle shows an overall decrease in apoptosis and an increase in cell survival. This type of cytoprotective mechanism could be aimed at preserving the existing cellular components during long-term anoxia and oxygen recovery phases in the wood frog.
Collapse
Affiliation(s)
- Victoria E M Gerber
- Department of Biology, Carleton University, Ottawa, ON, Canada; Current affiliation: Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | - Kenneth B Storey
- Institute of Biochemistry, Department of Biology and Chemistry, Carleton University , Ottawa, ON , Canada
| |
Collapse
|
21
|
Abdulkareem ZA, Gee JMW, Cox CD, Wann KT. Knockdown of the small conductance Ca(2+) -activated K(+) channels is potently cytotoxic in breast cancer cell lines. Br J Pharmacol 2016; 173:177-90. [PMID: 26454020 PMCID: PMC4737296 DOI: 10.1111/bph.13357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/27/2015] [Accepted: 09/24/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Small conductance calcium-activated potassium (KCa 2.x) channels have a widely accepted canonical function in regulating cellular excitability. In this study, we address a potential non-canonical function of KCa 2.x channels in breast cancer cell survival, using in vitro models. EXPERIMENTAL APPROACH The expression of all KCa 2.x channel isoforms was initially probed using RT-PCR, Western blotting and microarray analysis in five widely studied breast cancer cell lines. In order to assess the effect of pharmacological blockade and siRNA-mediated knockdown of KCa 2.x channels on these cell lines, we utilized MTS proliferation assays and also followed the corresponding expression of apoptotic markers. KEY RESULTS All of the breast cancer cell lines, regardless of their lineage or endocrine responsiveness, were highly sensitive to KCa 2.x channel blockade. UCL1684 caused cytotoxicity, with LD50 values in the low nanomolar range, in all cell lines. The role of KCa 2.x channels was confirmed using pharmacological inhibition and siRNA-mediated knockdown. This reduced cell viability and also reduced expression of Bcl-2 but increased expression of active caspase-7 and caspase-9. Complementary to these results, a variety of cell lines can be protected from apoptosis induced by staurosporine using the KCa 2.x channel activator CyPPA. CONCLUSIONS AND IMPLICATIONS In addition to a well-established role for KCa 2.x channels in migration, blockade of these channels was potently cytotoxic in breast cancer cell lines, pointing to modulation of KCa 2.x channels as a potential therapeutic approach to breast cancer.
Collapse
Affiliation(s)
| | - Julia MW Gee
- School of Pharmacy and Pharmaceutical SciencesCardiff UniversityCardiffCF10 3NBUK
| | - Charles D Cox
- Victor Chang Cardiac Research InstituteDarlinghurstNSW2010Australia
| | - Kenneth T Wann
- School of Pharmacy and Pharmaceutical SciencesCardiff UniversityCardiffCF10 3NBUK
| |
Collapse
|
22
|
Kapitsinou PP, Haase VH. Molecular mechanisms of ischemic preconditioning in the kidney. Am J Physiol Renal Physiol 2015; 309:F821-34. [DOI: 10.1152/ajprenal.00224.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/21/2015] [Indexed: 12/26/2022] Open
Abstract
More effective therapeutic strategies for the prevention and treatment of acute kidney injury (AKI) are needed to improve the high morbidity and mortality associated with this frequently encountered clinical condition. Ischemic and/or hypoxic preconditioning attenuates susceptibility to ischemic injury, which results from both oxygen and nutrient deprivation and accounts for most cases of AKI. While multiple signaling pathways have been implicated in renoprotection, this review will focus on oxygen-regulated cellular and molecular responses that enhance the kidney's tolerance to ischemia and promote renal repair. Central mediators of cellular adaptation to hypoxia are hypoxia-inducible factors (HIFs). HIFs play a crucial role in ischemic/hypoxic preconditioning through the reprogramming of cellular energy metabolism, and by coordinating adenosine and nitric oxide signaling with antiapoptotic, oxidative stress, and immune responses. The therapeutic potential of HIF activation for the treatment and prevention of ischemic injuries will be critically examined in this review.
Collapse
Affiliation(s)
- Pinelopi P. Kapitsinou
- Departments of Medicine, Anatomy and Cell Biology, and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Volker H. Haase
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
- Medicine and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
23
|
Downregulation of key regulatory proteins in androgen dependent prostate tumor cells by oncolytic reovirus. Virology 2015; 485:153-61. [PMID: 26264969 DOI: 10.1016/j.virol.2015.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 11/21/2022]
Abstract
As prostate tumor cell growth depends on hormones, androgen ablation is an effective therapy for prostate cancer (PCa). However, progression of PCa cells to androgen independent growth (castrate resistant prostate cancer, CRPC) results in relapse and mortality. Hypoxia, a microenvironment of low oxygen that modifies the activity of PCa regulatory proteins including the androgen receptor (AR), plays a critical role in progression to CRPC. Therapies targeting hypoxia and the AR may lengthen the time to CRPC progression thereby increasing survival time of PCa patients. Mammalian Orthoreovirus (MRV) has shown promise for the treatment of prostate tumors in vitro and in vivo. In this study, we found that MRV infection induces downregulation of proteins implicated in CRPC progression, interferes with hypoxia-induced AR activity, and induces apoptosis in androgen dependent cells. This suggests MRV possesses traits that could be exploited to create novel therapies for the inhibition of progression to CRPC.
Collapse
|
24
|
Khan S, Ferguson Bennit H, Asuncion Valenzuela MM, Turay D, Diaz Osterman CJ, Moyron RB, Esebanmen GE, Ashok A, Wall NR. Localization and upregulation of survivin in cancer health disparities: a clinical perspective. Biologics 2015; 9:57-67. [PMID: 26185415 PMCID: PMC4501680 DOI: 10.2147/btt.s83864] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Survivin is one of the most important members of the inhibitors of apoptosis protein family, as it is expressed in most human cancers but is absent in normal, differentiated tissues. Lending to its importance, survivin has proven associations with apoptosis and cell cycle control, and has more recently been shown to modulate the tumor microenvironment and immune evasion as a result of its extracellular localization. Upregulation of survivin has been found in many cancers including breast, prostate, pancreatic, and hematological malignancies, and it may prove to be associated with the advanced presentation, poorer prognosis, and lower survival rates observed in ethnically diverse populations.
Collapse
Affiliation(s)
- Salma Khan
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Heather Ferguson Bennit
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Malyn May Asuncion Valenzuela
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - David Turay
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Department of Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Carlos J Diaz Osterman
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Ron B Moyron
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Grace E Esebanmen
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Arjun Ashok
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Nathan R Wall
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA ; Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
25
|
Doktorova H, Hrabeta J, Khalil MA, Eckschlager T. Hypoxia-induced chemoresistance in cancer cells: The role of not only HIF-1. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:166-77. [PMID: 26001024 DOI: 10.5507/bp.2015.025] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/07/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The aim of this review is to provide the information about molecular basis of hypoxia-induced chemoresistance, focusing on the possibility of diagnostic and therapeutic use. RESULTS Hypoxia is a common feature of tumors and represents an independent prognostic factor in many cancers. It is the result of imbalances in the intake and consumption of oxygen caused by abnormal vessels in the tumor and the rapid proliferation of cancer cells. Hypoxia-induced resistance to cisplatin, doxorubicin, etoposide, melphalan, 5-flouoruracil, gemcitabine, and docetaxel has been reported in a number of experiments. Adaptation of tumor cells to hypoxia has important biological effects. The most studied factor responsible for these effects is hypoxia-inducible factor-1 (HIF-1) that significantly contributes to the aggressiveness and chemoresistance of different tumors. The HIF-1 complex, induced by hypoxia, binds to target genes, thereby increasing the expression of many genes. In addition, the expression of hundreds of genes can be also decreased in response to hypoxia in HIF-1 dependent manner, but without the detection of HIF-1 in these genes' promoters. HIF-1 independent mechanisms for drug resistance in hypoxia have been described, however, they are still rarely reported. The first clinical studies focusing on diagnosis of hypoxia and on inhibition of hypoxia-induced changes in cancer cells are starting to yield results. CONCLUSIONS The adaptation to hypoxia requires many genetic and biochemical responses that regulate one another. Hypoxia-induced resistance is a very complex field and we still know very little about it. Different approaches to circumvent hypoxia in tumors are under development.
Collapse
Affiliation(s)
- Helena Doktorova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Jan Hrabeta
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Mohamed Ashraf Khalil
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
26
|
Smeyne M, Sladen P, Jiao Y, Dragatsis I, Smeyne RJ. HIF1α is necessary for exercise-induced neuroprotection while HIF2α is needed for dopaminergic neuron survival in the substantia nigra pars compacta. Neuroscience 2015; 295:23-38. [PMID: 25796140 DOI: 10.1016/j.neuroscience.2015.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/05/2015] [Accepted: 03/07/2015] [Indexed: 02/05/2023]
Abstract
Exercise reduces the risk of developing a number of neurological disorders and increases the efficiency of cellular energy production. However, overly strenuous exercise produces oxidative stress. Proper oxygenation is crucial for the health of all tissues, and tight regulation of cellular oxygen is critical to balance O2 levels and redox homeostasis in the brain. Hypoxia Inducible Factor (HIF)1α and HIF2α are transcription factors regulated by cellular oxygen concentration that initiate gene regulation of vascular development, redox homeostasis, and cell cycle control. HIF1α and HIF2α contribute to important adaptive mechanisms that occur when oxygen and ROS homeostasis become unbalanced. It has been shown that preconditioning by exposure to a stressor prior to a hypoxic event reduces damage that would otherwise occur. Previously we reported that 3 months of exercise protects SNpc dopaminergic (DA) neurons from toxicity caused by Complex I inhibition. Here, we identify the cells in the SNpc that express HIF1α and HIF2α and show that running exercise produces hypoxia in SNpc DA neurons, and alters the expression of HIF1α and HIF2α. In mice carrying a conditional knockout of Hif1α in postnatal neurons we observe that exercise alone produces SNpc TH+ DA neuron loss. Loss of HIF1α also abolishes exercise-induced neuroprotection. In mice lacking Hif2α in postnatal neurons, the number of TH+ DA neurons in the adult SNpc is diminished, but 3months of exercise rescues this loss. We conclude that HIF1α is necessary for exercise-induced neuroprotection and both HIF1α and HIF2α are necessary for the survival and function of adult SNpc DA neurons.
Collapse
Affiliation(s)
- M Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - P Sladen
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - Y Jiao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - I Dragatsis
- Department of Physiology, The University of Tennessee Health Science Center, 894 Union Avenue, Memphis, TN 38163, United States
| | - R J Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States.
| |
Collapse
|
27
|
Gupta-Saraf P, Miller CL. HIF-1α downregulation and apoptosis in hypoxic prostate tumor cells infected with oncolytic mammalian orthoreovirus. Oncotarget 2015; 5:561-74. [PMID: 24504474 PMCID: PMC3964229 DOI: 10.18632/oncotarget.1767] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypoxia has emerged as one of the most important drivers of tumor aggression, metastasis, and poor clinical outcome in many cancers.In prostate cancer (PCa), hypoxia has been strongly correlated to biochemical failure and local recurrence. However, current PCa treatment options do not address hypoxic cells highlighting a critical gap in existing therapies and the need for development of therapies that target hypoxic prostate tumor cells. Mammalian orthoreovirus (MRV) is an oncolytic virus that targets tumor cells over normal cells which has been shown to be safe and effective against many cancers in vitro, in animal models, and in human clinical trials. We found that MRVinfects and replicates in hypoxic prostate tumor cells to levels comparable to normoxic cells leading to apoptosis and cell death. In addition, the regulatory subunit (HIF-1α) of the master transcriptional regulator of hypoxia, HIF-1, was significantly downregulated in infected cells. HIF-1α downregulation was found to occur via ubiquitin-dependent proteasome-mediated degradation and translational inhibition. Virus-mediated HIF-1α degradation required the HIF-1α PAS domain and expression of the receptor for activated kinase C (RACK1) protein. These data provide evidence that MRV may be a viable therapeutic option for targeting hypoxic cells and HIF-1α in PCa.
Collapse
Affiliation(s)
- Pooja Gupta-Saraf
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA
| | | |
Collapse
|
28
|
Hsieh CH, Lin YJ, Wu CP, Lee HT, Shyu WC, Wang CC. Livin contributes to tumor hypoxia-induced resistance to cytotoxic therapies in glioblastoma multiforme. Clin Cancer Res 2014; 21:460-70. [PMID: 25370472 DOI: 10.1158/1078-0432.ccr-14-0618] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor hypoxia is one of the crucial microenvironments to promote therapy resistance (TR) in glioblastoma multiforme (GBM). Livin, a member of the family of inhibitor of apoptosis proteins, contributes antiapoptosis. However, the role of tumor hypoxia in Livin regulation and its impact on TR are unclear. EXPERIMENTAL DESIGN Livin expression and apoptosis for tumor hypoxic cells derived from human glioblastoma xenografts or in vitro hypoxic stress-treated glioblastoma cells were determined by Western blotting, immunofluorescence imaging, and annexin V staining assay. The mechanism of hypoxia-induced Livin induction was investigated by chromatin immunoprecipitation assay and reporter assay. Genetic and pharmacologic manipulation of Livin was utilized to investigate the role of Livin on tumor hypoxia-induced TR in vitro or in vivo. RESULTS The upregulation of Livin expression and downregulation of caspase activity were observed under cycling and chronic hypoxia in glioblastoma cells and xenografts, concomitant with increased TR to ionizing radiation and temozolomide. However, knockdown of Livin inhibited these effects. Moreover, hypoxia activated Livin transcription through the binding of hypoxia-inducible factor-1α to the Livin promoter. The targeted inhibition of Livin by the cell-permeable peptide (TAT-Lp15) in intracerebral glioblastoma-bearing mice demonstrated a synergistic suppression of tumor growth and increased the survival rate in standard-of-care treatment with radiation plus temozolomide. CONCLUSIONS These findings indicate a novel pathway that links upregulation of Livin to tumor hypoxia-induced TR in GBM and suggest that targeting Livin using cell-permeable peptide may be an effective therapeutic strategy for tumor microenvironment-induced TR.
Collapse
Affiliation(s)
- Chia-Hung Hsieh
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan. Department of Medical Research, China Medical University Hospital, Taichung, Taiwan. Department of Biomedical Informatics, Asia University, Taichung, Taiwan.
| | - Yu-Jung Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chung-Pu Wu
- Department of Physiology and Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Department of Neurology, Center for Neuropsychiatry, China Medical University and Hospital, Taichung, Taiwan. Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.
| | - Chi-Chung Wang
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei, Taiwan.
| |
Collapse
|
29
|
Clarke RH, Moosa S, Anzivino M, Wang Y, Floyd DH, Purow BW, Lee KS. Sustained radiosensitization of hypoxic glioma cells after oxygen pretreatment in an animal model of glioblastoma and in vitro models of tumor hypoxia. PLoS One 2014; 9:e111199. [PMID: 25350400 PMCID: PMC4211739 DOI: 10.1371/journal.pone.0111199] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/29/2014] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal form of brain cancer and these tumors are highly resistant to chemo- and radiotherapy. Radioresistance is thought to result from a paucity of molecular oxygen in hypoxic tumor regions, resulting in reduced DNA damage and enhanced cellular defense mechanisms. Efforts to counteract tumor hypoxia during radiotherapy are limited by an attendant increase in the sensitivity of healthy brain tissue to radiation. However, the presence of heightened levels of molecular oxygen during radiotherapy, while conventionally deemed critical for adjuvant oxygen therapy to sensitize hypoxic tumor tissue, might not actually be necessary. We evaluated the concept that pre-treating tumor tissue by transiently elevating tissue oxygenation prior to radiation exposure could increase the efficacy of radiotherapy, even when radiotherapy is administered after the return of tumor tissue oxygen to hypoxic baseline levels. Using nude mice bearing intracranial U87-luciferase xenografts, and in vitro models of tumor hypoxia, the efficacy of oxygen pretreatment for producing radiosensitization was tested. Oxygen-induced radiosensitization of tumor tissue was observed in GBM xenografts, as seen by suppression of tumor growth and increased survival. Additionally, rodent and human glioma cells, and human glioma stem cells, exhibited prolonged enhanced vulnerability to radiation after oxygen pretreatment in vitro, even when radiation was delivered under hypoxic conditions. Over-expression of HIF-1α reduced this radiosensitization, indicating that this effect is mediated, in part, via a change in HIF-1-dependent mechanisms. Importantly, an identical duration of transient hyperoxic exposure does not sensitize normal human astrocytes to radiation in vitro. Taken together, these results indicate that briefly pre-treating tumors with elevated levels of oxygen prior to radiotherapy may represent a means for selectively targeting radiation-resistant hypoxic cancer cells, and could serve as a safe and effective adjuvant to radiation therapy for patients with GBM.
Collapse
Affiliation(s)
- Ryon H. Clarke
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Shayan Moosa
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Matthew Anzivino
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Yi Wang
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Desiree Hunt Floyd
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Benjamin W. Purow
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Kevin S. Lee
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States of America
| |
Collapse
|
30
|
Zhang YB, Wang X, Meister EA, Gong KR, Yan SC, Lu GW, Ji XM, Shao G. The effects of CoCl2 on HIF-1α protein under experimental conditions of autoprogressive hypoxia using mouse models. Int J Mol Sci 2014; 15:10999-1012. [PMID: 24945310 PMCID: PMC4100194 DOI: 10.3390/ijms150610999] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/22/2022] Open
Abstract
It is well known that cobalt chloride (CoCl2) can enhance the stability of hypoxia-inducible factor (HIF)-1α. The aim of this study is to detect the effect of CoCl2 on the hypoxia tolerance of mice which were repeatedly exposed to autoprogressive hypoxia. Balb/c mice were randomly divided into groups of chemical pretreatment and normal saline (NS), respectively injected with CoCl2 and NS 3 h before exposure to hypoxia for 0 run (H0), 1 run (H1), and 4 runs (H4). Western Blot, electrophoretic mobility shift assay (EMSA), extracellular recordings population spikes in area cornus ammonis I (CA 1) of mouse hippocampal slices and real-time were used in this study. Our results demonstrated that the tolerance of mice to hypoxia, the changes of HIF-1α protein level and HIF-1 DNA binding activity in mice hippocampus, the mRNA level of erythropoietin (EPO) and vascular endothelial growth factor (VEGF), and the disappearance time of population spikes of hippocampal slices were substantially different between the control group and the CoCl2 group. Over-induction of HIF-1α by pretreatment with CoCl2 before hypoxia did not increase the hypoxia tolerance.
Collapse
Affiliation(s)
- Yan-Bo Zhang
- Department of Neurology, Affiliated Hospital of Tai Shan Medical University, Taishan 271000, China.
| | - Xiulian Wang
- Department of Intensive Care Unit , 2nd Affiliated Hospital of Baotou Medical College, Baotou 014030, China.
| | - Edward A Meister
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Ke-Rui Gong
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Shao-Chun Yan
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Guo-Wei Lu
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Xun-Ming Ji
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Guo Shao
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| |
Collapse
|
31
|
Zhou Z, Liu F, Zhang ZS, Shu F, Zheng Y, Fu L, Li LY. Human rhomboid family-1 suppresses oxygen-independent degradation of hypoxia-inducible factor-1α in breast cancer. Cancer Res 2014; 74:2719-30. [PMID: 24648344 DOI: 10.1158/0008-5472.can-13-1027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intermittent oxygen deficiency in cancers promotes prolonged inflammation, continuous angiogenesis, and increased drug resistance. Hypoxia-inducible factor-1 (HIF1) has a pivotal role in the regulation of cellular responses to oxygen deficiency. The α-subunit of HIF1 (HIF1α) is degraded in normoxia but stabilized in hypoxia. However, the molecular mechanism that controls oxygen-independent degradation of HIF1α has remained elusive. Human rhomboid family-1 (RHBDF1) is a member of a large family of nonprotease rhomboids whose function is basically unknown. We report here that RHBDF1 expression in breast cancer is highly elevated and is strongly correlated with escalated disease progression, metastasis, poor prognosis, and poor response to chemotherapy. We show that RHBDF1 interaction with the receptor of activated protein-C kinase-1 (RACK1) in breast cancer cells prevents RACK1-assisted, oxygen-independent HIF1α degradation. In addition, we show that the HIF1α-stabilizing activity of RHBDF1 diminishes when the phosphorylation of a tyrosine residue on the RHBDF1 molecule is inhibited. These findings are consistent with the view that RHBDF1 is a critical component of a molecular switch that regulates HIF1α stability in cancer cells in hypoxia and that RHBDF1 is of potential value as a new target for cancer treatment.
Collapse
Affiliation(s)
- Zhuan Zhou
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Fangfang Liu
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhi-Song Zhang
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Feifei Shu
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yangyang Zheng
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Li Fu
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lu-Yuan Li
- Authors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, ChinaAuthors' Affiliations: State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University; Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; and Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
32
|
Yang W, Cooke M, Duckett CS, Yang X, Dorsey JF. Distinctive effects of the cellular inhibitor of apoptosis protein c-IAP2 through stabilization by XIAP in glioblastoma multiforme cells. Cell Cycle 2014; 13:992-1005. [PMID: 24552816 DOI: 10.4161/cc.27880] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inhibitor of apoptosis proteins (IAPs) are extensively involved in NFκB signaling pathways. Regulation of c-IAP2 turnover by other proteins was investigated in glioblastoma multiforme (GBM) cells in the present study. When overexpressed, X-linked IAP (XIAP) enhanced expression of ectopic c-IAP2, but not c-IAP1, and endogenous c-IAP2 levels were reduced once XIAP expression was silenced. TNFα stimulation substantially increased c-IAP2 expression, and this upregulation was impaired by suppression of XIAP. Similarly, when XIAP was limiting due to severe hypoxic conditions, c-IAP2 levels were downregulated. These data together indicate that XIAP is an important regulator responsible for stabilization of c-IAP2 levels under different conditions. Protein interactions occur through binding of BIR2 and BIR3 domains of c-IAP2 with the RING finger of XIAP. XIAP inhibition of c-IAP2 auto-degradation was dependent on this physical interaction, and it was independent of XIAP E3 ligase activity. Global c-IAP2 ubiquitination was not affected by XIAP, although c-IAP2 levels were significantly increased. A CARD-RING-containing fragment of c-IAP2 was found to target XIAP for proteasome-independent degradation, but it was unable to sensitize GBM cells to chemo-reagents. The XIAP-stabilized c-IAP2 was found to enhance IκB-α phosphorylation on serines 32 and 36, and to antagonize XIAP-induced increase in mature Smac and Bcl10. Taken together, our data identify a distinctive role of c-IAP2 as stabilizer of XIAP, which is likely involved in regulation of NFκB activation and apoptosis in GBM cells.
Collapse
Affiliation(s)
- Wensheng Yang
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Mariana Cooke
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Colin S Duckett
- Departments of Pathology and Internal Medicine and Comprehensive Cancer Center; University of Michigan; Ann Arbor, MI USA
| | - Xiaolu Yang
- Department of Cancer Biology and the Abramson Family Cancer Research Institute; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Jay F Dorsey
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|
33
|
Glazer PM, Hegan DC, Lu Y, Czochor J, Scanlon SE. Hypoxia and DNA repair. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2013; 86:443-51. [PMID: 24348208 PMCID: PMC3848098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hypoxia is a characteristic feature of solid tumors and occurs very early in neoplastic development. Hypoxia transforms cell physiology in multiple ways, with profound changes in cell metabolism, cell growth, susceptibility to apoptosis, induction of angiogenesis, and increased motility. Over the past 20 years, our lab has determined that hypoxia also induces genetic instability. We have conducted a large series of experiments revealing that this instability occurs through the alteration of DNA repair pathways, including nucleotide excision repair, DNA mismatch repair, and homology dependent repair. Our work suggests that hypoxia, as a key component of solid tumors, can drive cancer progression through its impact on genomic integrity. However, the acquired changes in DNA repair that are induced by hypoxia may also render hypoxic cancer cells vulnerable to tailored strategies designed to exploit these changes.
Collapse
Affiliation(s)
- Peter M. Glazer
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut,To whom all correspondence should be
addressed: Peter M. Glazer, Department of Therapeutic Radiology, Yale
University, 333 Cedar St., New Haven, CT 06520-8040; Tele: 203-737-2788; Fax:
203-737-1467;
| | - Denise C. Hegan
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut
| | - Yuhong Lu
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut
| | - Jennifer Czochor
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut
| | - Susan E. Scanlon
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut
| |
Collapse
|
34
|
Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3481-3498. [PMID: 23830918 DOI: 10.1016/j.bbamcr.2013.06.026] [Citation(s) in RCA: 808] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/21/2013] [Accepted: 06/22/2013] [Indexed: 02/07/2023]
Abstract
Anoikis is a programmed cell death induced upon cell detachment from extracellular matrix, behaving as a critical mechanism in preventing adherent-independent cell growth and attachment to an inappropriate matrix, thus avoiding colonizing of distant organs. As anchorage-independent growth and epithelial-mesenchymal transition, two features associated with anoikis resistance, are vital steps during cancer progression and metastatic colonization, the ability of cancer cells to resist anoikis has now attracted main attention from the scientific community. Cancer cells develop anoikis resistance due to several mechanisms, including change in integrins' repertoire allowing them to grow in different niches, activation of a plethora of inside-out pro-survival signals as over-activation of receptors due to sustained autocrine loops, oncogene activation, growth factor receptor overexpression, or mutation/upregulation of key enzymes involved in integrin or growth factor receptor signaling. In addition, tumor microenvironment has also been acknowledged to contribute to anoikis resistance of bystander cancer cells, by modulating matrix stiffness, enhancing oxidative stress, producing pro-survival soluble factors, triggering epithelial-mesenchymal transition and self-renewal ability, as well as leading to metabolic deregulations of cancer cells. All these events help cancer cells to inhibit the apoptosis machinery and sustain pro-survival signals after detachment, counteracting anoikis and constituting promising targets for anti-metastatic pharmacological therapy. This article is part of a Special Section entitled: Cell Death Pathways.
Collapse
Affiliation(s)
- Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; Tuscany Tumor Institute and "Center for Research, Transfer and High Education, DENOTHE", 50134 Florence, Italy.
| |
Collapse
|
35
|
Adamski J, Price A, Dive C, Makin G. Hypoxia-induced cytotoxic drug resistance in osteosarcoma is independent of HIF-1Alpha. PLoS One 2013; 8:e65304. [PMID: 23785417 PMCID: PMC3681794 DOI: 10.1371/journal.pone.0065304] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 04/24/2013] [Indexed: 01/06/2023] Open
Abstract
Survival rates from childhood cancer have improved dramatically in the last 40 years, such that over 80% of children are now cured. However in certain subgroups, including metastatic osteosarcoma, survival has remained stubbornly poor, despite dose intensive multi-agent chemotherapy regimens, and new therapeutic approaches are needed. Hypoxia is common in adult solid tumours and is associated with treatment resistance and poorer outcome. Hypoxia induces chemotherapy resistance in paediatric tumours including neuroblastoma, rhabdomyosarcoma and Ewing’s sarcoma, in vitro, and this drug resistance is dependent on the oxygen-regulated transcription factor hypoxia inducible factor-1 (HIF-1). In this study the effects of hypoxia on the response of the osteosarcoma cell lines 791T, HOS and U2OS to the clinically relevant cytotoxics cisplatin, doxorubicin and etoposide were evaluated. Significant hypoxia-induced resistance to all three agents was seen in all three cell lines and hypoxia significantly reduced drug-induced apoptosis. Hypoxia also attenuated drug-induced activation of p53 in the p53 wild-type U2OS osteosarcoma cells. Drug resistance was not induced by HIF-1α stabilisation in normoxia by cobalt chloride nor reversed by the suppression of HIF-1α in hypoxia by shRNAi, siRNA, dominant negative HIF or inhibition with the small molecule NSC-134754, strongly suggesting that hypoxia-induced drug resistance in osteosarcoma cells is independent of HIF-1α. Inhibition of the phosphoinositide 3-kinase (PI3K) pathway using the inhibitor PI-103 did not reverse hypoxia-induced drug resistance, suggesting the hypoxic activation of Akt in osteosarcoma cells does not play a significant role in hypoxia-induced drug resistance. Targeting hypoxia is an exciting prospect to improve current anti-cancer therapy and combat drug resistance. Significant hypoxia-induced drug resistance in osteosarcoma cells highlights the potential importance of hypoxia as a target to reverse drug resistance in paediatric osteosarcoma. The novel finding of HIF-1α independent drug resistance suggests however other hypoxia related targets may be more relevant in paediatric osteosarcoma.
Collapse
Affiliation(s)
- Jennifer Adamski
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, Manchester, United Kingdom
- Institute of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Department of Paediatric Oncology, Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | - Andrew Price
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, Manchester, United Kingdom
- Institute of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Guy Makin
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, Manchester, United Kingdom
- Institute of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Department of Paediatric Oncology, Royal Manchester Children’s Hospital, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Iranon NN, Miller DL. Interactions between oxygen homeostasis, food availability, and hydrogen sulfide signaling. Front Genet 2012; 3:257. [PMID: 23233860 PMCID: PMC3516179 DOI: 10.3389/fgene.2012.00257] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/04/2012] [Indexed: 12/19/2022] Open
Abstract
The ability to sense and respond to stressful conditions is essential to maintain organismal homeostasis. It has long been recognized that stress response factors that improve survival in changing conditions can also influence longevity. In this review, we discuss different strategies used by animals in response to decreased O(2) (hypoxia) to maintain O(2) homeostasis, and consider interactions between hypoxia responses, nutritional status, and H(2)S signaling. O(2) is an essential environmental nutrient for almost all metazoans as it plays a fundamental role in development and cellular metabolism. However, the physiological response(s) to hypoxia depend greatly on the amount of O(2) available. Animals must sense declining O(2) availability to coordinate fundamental metabolic and signaling pathways. It is not surprising that factors involved in the response to hypoxia are also involved in responding to other key environmental signals, particularly food availability. Recent studies in mammals have also shown that the small gaseous signaling molecule hydrogen sulfide (H(2)S) protects against cellular damage and death in hypoxia. These results suggest that H(2)S signaling also integrates with hypoxia response(s). Many of the signaling pathways that mediate the effects of hypoxia, food deprivation, and H(2)S signaling have also been implicated in the control of lifespan. Understanding how these pathways are coordinated therefore has the potential to reveal new cellular and organismal homeostatic mechanisms that contribute to longevity assurance in animals.
Collapse
Affiliation(s)
- Nicole N Iranon
- Department of Biochemistry, University of Washington School of Medicine Seattle, WA, USA ; Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine Seattle, WA, USA
| | | |
Collapse
|
37
|
Cimino F, Balestra C, Germonpré P, De Bels D, Tillmans F, Saija A, Speciale A, Virgili F. Pulsed high oxygen induces a hypoxic-like response in human umbilical endothelial cells and in humans. J Appl Physiol (1985) 2012; 113:1684-9. [PMID: 23042909 DOI: 10.1152/japplphysiol.00922.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
It has been proposed that relative changes of oxygen availability, rather than steady-state hypoxic or hyperoxic conditions, play an important role in hypoxia-inducible factor (HIF) transcriptional effects. According to this hypothesis describing the "normobaric oxygen paradox", normoxia following a hyperoxic event is sensed by tissues as an oxygen shortage, upregulating HIF-1 activity. With the aim of confirming, at cellular and at functional level, that normoxia following a hyperoxic event is "interpreted" as a hypoxic event, we report a combination of experiments addressing the effects of an intermittent increase of oxygen concentration on HIF-1 levels and the activity level of specific oxygen-modulated proteins in cultured human umbilical vein endothelial cells and the effects of hemoglobin levels after intermittent breathing of normobaric high (100%) and low (15%) oxygen in vivo in humans. Our experiments confirm that, during recovery after hyperoxia, an increase of HIF expression occurs in human umbilical vein endothelial cells, associated with an increase of matrix metalloproteinases activity. These data suggest that endothelial cells "interpret" the return to normoxia after hyperoxia as a hypoxic stimulus. At functional level, our data show that breathing both 15 and 100% oxygen 30 min every other day for a period of 10 days induces an increase of hemoglobin levels in humans. This effect was enhanced after the cessation of the oxygen breathing. These results indicate that a sudden decrease in tissue oxygen tension after hyperoxia may act as a trigger for erythropoietin synthesis, thus corroborating the hypothesis that "relative" hypoxia is a potent stimulator of HIF-mediated gene expressions.
Collapse
Affiliation(s)
- F Cimino
- Department Farmaco-Biologico, School of Pharmacy, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Prabhakar NR, Semenza GL. Adaptive and maladaptive cardiorespiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2. Physiol Rev 2012; 92:967-1003. [PMID: 22811423 DOI: 10.1152/physrev.00030.2011] [Citation(s) in RCA: 478] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is a fundamental stimulus that impacts cells, tissues, organs, and physiological systems. The discovery of hypoxia-inducible factor-1 (HIF-1) and subsequent identification of other members of the HIF family of transcriptional activators has provided insight into the molecular underpinnings of oxygen homeostasis. This review focuses on the mechanisms of HIF activation and their roles in physiological and pathophysiological responses to hypoxia, with an emphasis on the cardiorespiratory systems. HIFs are heterodimers comprised of an O(2)-regulated HIF-1α or HIF-2α subunit and a constitutively expressed HIF-1β subunit. Induction of HIF activity under conditions of reduced O(2) availability requires stabilization of HIF-1α and HIF-2α due to reduced prolyl hydroxylation, dimerization with HIF-1β, and interaction with coactivators due to decreased asparaginyl hydroxylation. Stimuli other than hypoxia, such as nitric oxide and reactive oxygen species, can also activate HIFs. HIF-1 and HIF-2 are essential for acute O(2) sensing by the carotid body, and their coordinated transcriptional activation is critical for physiological adaptations to chronic hypoxia including erythropoiesis, vascularization, metabolic reprogramming, and ventilatory acclimatization. In contrast, intermittent hypoxia, which occurs in association with sleep-disordered breathing, results in an imbalance between HIF-1α and HIF-2α that causes oxidative stress, leading to cardiorespiratory pathology.
Collapse
Affiliation(s)
- Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, Biological Sciences Division, University of Chicago, Chicago, Illinois, USA.
| | | |
Collapse
|
39
|
Xiao L, Kovac S, Chang M, Shulkes A, Baldwin GS, Patel O. Induction of gastrin expression in gastrointestinal cells by hypoxia or cobalt is independent of hypoxia-inducible factor (HIF). Endocrinology 2012; 153:3006-16. [PMID: 22593272 PMCID: PMC3380302 DOI: 10.1210/en.2011-2069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gastrin and its precursors have been shown to promote mitogenesis and angiogenesis in gastrointestinal tumors. Hypoxia stimulates tumor growth, but its effect on gastrin gene regulation has not been examined in detail. Here we have investigated the effect of hypoxia on the transcription of the gastrin gene in human gastric cancer (AGS) cells. Gastrin mRNA was measured by real-time PCR, gastrin peptides were measured by RIA, and gastrin promoter activity was measured by dual-luciferase reporter assay. Exposure to a low oxygen concentration (1%) increased gastrin mRNA concentrations in wild-type AGS cells (AGS) and in AGS cells overexpressing the gastrin receptor (AGS-cholecystokinin receptor 2) by 2.1 ± 0.4- and 4.1 ± 0.3-fold (P < 0.05), respectively. The hypoxia mimetic, cobalt chloride (300 μM), increased gastrin promoter activity in AGS cells by 2.4 ± 0.3-fold (P < 0.05), and in AGS-cholecystokinin receptor 2 cells by 4.0 ± 0.3-fold (P < 0.05), respectively. The observations that either deletion from the gastrin promoter of the putative binding sites for the transcription factor hypoxia-inducible factor 1 (HIF-1) or knockdown of either the HIF-1α or HIF-1β subunit did not affect gastrin promoter inducibility under hypoxia indicated that the hypoxic activation of the gastrin gene is likely HIF independent. Mutational analysis of previously identified Sp1 regulatory elements in the gastrin promoter also failed to abrogate the induction of promoter activity by hypoxia. The observations that hypoxia up-regulates the gastrin gene in AGS cells by HIF-independent mechanisms, and that this effect is enhanced by the presence of gastrin receptors, provide potential targets for gastrointestinal cancer therapy.
Collapse
Affiliation(s)
- Lin Xiao
- The University of Melbourne, Department of Surgery, Austin Health, Studley Road, Heidelberg, Victoria 3084, Australia
| | | | | | | | | | | |
Collapse
|
40
|
Zhao N, Sun BC, Sun T, Ma YM, Zhao XL, Liu ZY, Dong XY, Che N, Mo J, Gu Q. Hypoxia-induced vasculogenic mimicry formation via VE-cadherin regulation by Bcl-2. Med Oncol 2012; 29:3599-607. [PMID: 22562824 DOI: 10.1007/s12032-012-0245-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 04/18/2012] [Indexed: 12/21/2022]
Abstract
Vasculogenic mimicry (VM) refers to the unique ability of highly aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks. Hypoxia plays a pivotal role in the formation of VM. Hypoxia-induced Bcl-2 overexpression is observed in many types of tumors including melanoma, in which it is associated with tumorigenicity and angiogenesis. VE-cadherin, the major endothelial adhesion molecule controlling cellular junctions and blood vessel formation, is also overexpressed in melanoma. Despite these connections, whether hypoxia induces VM formation via VE-cadherin regulation by Bcl-2 is not confirmed. We used human melanoma cells to upregulate or knockdown the expression of Bcl-2 to investigate the possible molecular mechanism of VM formation under hypoxia. Bcl-2 overexpression increased VE-cadherin expression and VM formation under normoxia, whereas Bcl-2 siRNA significantly decreased VE-cadherin expression and VM formation under hypoxia. We then demonstrated that Bcl-2 regulated VE-cadherin transcription activity by Western blot, three-dimensional cultures, reporter gene assay, and clinical analysis. Therefore, Bcl-2-dependent VE-cadherin overexpression may be an important mechanism by which hypoxia induces VM.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pathology, Tianjin Medical University, Heping District Qixiangtai Road No. 22, Tianjin, 300070, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Kim YA, Chang M, Park YJ, Kim JE. Detection of Survivin and COX-2 in Thyroid Carcinoma: Anaplastic Carcinoma Shows Overexpression of Nuclear Survivin and Low COX-2 Expression. KOREAN JOURNAL OF PATHOLOGY 2012; 46:55-60. [PMID: 23109979 PMCID: PMC3479706 DOI: 10.4132/koreanjpathol.2012.46.1.55] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/09/2012] [Accepted: 01/10/2012] [Indexed: 11/17/2022]
Abstract
BACKGROUND Overexpression of survivin, a member of the inhibitors of apoptosis protein, has been reported in various carcinomas, and its interaction with cyclooxygenase 2 (COX-2) results in accelerated tumor progression. The purpose of this study is to investigate the immunohistochemical expression of survivin and COX-2 in benign and malignant thyroid tissues and to define its association with pathologic and clinical features. METHODS We examined expression of survivin and COX-2 by immunohistochemistry in 334 benign and malignant thyroid tissues and evaluated their clinical significance. RESULTS Expression of survivin showed an increase along the spectrum of thyroid carcinoma progression; rarely positive in adenomatous goiter, moderately positive in papillary carcinoma, and strongly positive in anaplastic carcinoma (AC). Papillary microcarcinoma revealed the highest COX-2 positivity and AC demonstrated the lowest positivity among thyroid cancers. Node negative carcinomas showed higher COX-2 expression than node positive tumors. Survivin expression did not correlate with COX-2. CONCLUSIONS Our findings suggest that survivin overexpression may be related to the pathogenesis of AC and can be a predictor of disease progression. COX-2 may be involved in the early phase of thyroid carcinoma.
Collapse
Affiliation(s)
- Young A Kim
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
43
|
Cipro Š, Hřebačková J, Hraběta J, Poljaková J, Eckschlager T. Valproic acid overcomes hypoxia-induced resistance to apoptosis. Oncol Rep 2011; 27:1219-26. [PMID: 22159638 PMCID: PMC3583540 DOI: 10.3892/or.2011.1577] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 11/17/2011] [Indexed: 01/12/2023] Open
Abstract
Valproic acid (VPA), a histone deacetylase inhibitor (HDACi), has been shown to be an effective tool in cancer treatment. Although its ability to induce apoptosis has been described in many cancer types, the data come from experiments performed in normoxic (21% O2) conditions only. Therefore, we questioned whether VPA would be equally effective under hypoxic conditions (1% O2), which is known to induce resistance to apoptosis. Four neuroblastoma cell lines were used: UKF-NB-3, SK-N-AS, plus one cisplatin-resistant subline derived from each of the two original sensitive lines. All were treated with VPA and incubated under hypoxic conditions. Measurement of apoptosis and viability using TUNEL assay and Annexin V/propidium iodide labeling revealed that VPA was even more effective under hypoxic conditions. We show here that hypoxia-induced resistance to chemotherapeutic agents such as cisplatin could be overcome using VPA. We also demonstrated that apoptosis pathways induced by VPA do not differ between normoxic and hypoxic conditions. VPA-induced apoptosis proceeds through the mitochondrial pathway, not the extrinsic pathway (under both normoxia and hypoxia), since inhibition of caspase-8 failed to decrease apoptosis or influence bid cleavage. Our data demonstrated that VPA is more efficient in triggering apoptosis under hypoxic conditions and overcomes hypoxia-induced resistance to cisplatin. The results provide additional evidence for the use of VPA in neuroblastoma (NBL) treatment.
Collapse
Affiliation(s)
- Šimon Cipro
- Department of Pediatric Hematology and Oncology, 2nd Medical School, Charles University and University Hospital Motol, V Úvalu 84, 150 00 Prague 5, Czech Republic
| | | | | | | | | |
Collapse
|
44
|
Kim Y, Jang M, Lim S, Won H, Yoon KS, Park JH, Kim HJ, Kim BH, Park WS, Ha J, Kim SS. Role of cyclophilin B in tumorigenesis and cisplatin resistance in hepatocellular carcinoma in humans. Hepatology 2011; 54:1661-1678. [PMID: 21748762 DOI: 10.1002/hep.24539] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Cyclophilin B (CypB) performs diverse roles in living cells, but its role in hepatocellular carcinoma (HCC) is largely unclear. To reveal its role in HCC, we investigated the induction of CypB under hypoxia and its functions in tumor cells in vitro and in vivo. Here, we demonstrated that hypoxia-inducible factor 1α (HIF-1α) induces CypB under hypoxia. Interestingly, CypB protected tumor cells, even p53-defective HCC cells, against hypoxia- and cisplatin-induced apoptosis. Furthermore, it regulated the effects of HIF-1α, including those in angiogenesis and glucose metabolism, via a positive feedback loop with HIF-1α. The tumorigenic and chemoresistant effects of CypB were confirmed in vivo using a xenograft model. Finally, we showed that CypB is overexpressed in 78% and 91% of the human HCC and colon cancer tissues, respectively, and its overexpression in these cancers reduced patient survival. CONCLUSIONS These results indicate that CypB induced by hypoxia stimulates the survival of HCC via a positive feedback loop with HIF-1α, indicating that CypB is a novel candidate target for developing chemotherapeutic agents against HCC and colon cancer.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cisplatin/pharmacology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Cyclophilins/genetics
- Cyclophilins/metabolism
- Drug Resistance, Neoplasm/physiology
- Female
- Hep G2 Cells
- Humans
- Hydrogen Peroxide/pharmacology
- Hypoxia/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Oxidants/pharmacology
- Promoter Regions, Genetic/physiology
- STAT3 Transcription Factor/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yeonghwan Kim
- Department of Biochemistry and Molecular Biology (BK21 project), Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mayes PA, Dolloff NG, Daniel CJ, Liu JJ, Hart LS, Kuribayashi K, Allen JE, Jee DI, Dorsey JF, Liu YY, Dicker DT, Brown JM, Furth EE, Klein PS, Sears RC, El-Deiry WS. Overcoming hypoxia-induced apoptotic resistance through combinatorial inhibition of GSK-3β and CDK1. Cancer Res 2011; 71:5265-75. [PMID: 21646472 PMCID: PMC3667402 DOI: 10.1158/0008-5472.can-11-1383] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor hypoxia is an inherent impediment to cancer treatment that is both clinically significant and problematic. In this study, we conducted a cell-based screen to identify small molecules that could reverse the apoptotic resistance of hypoxic cancer cells. Among the compounds, we identified were a structurally related group that sensitized hypoxic cancer cells to apoptosis by inhibiting the kinases GSK-3β and cyclin-dependent kinase (CDK) 1. Combinatorial inhibition of these proteins in hypoxic cancer cells and tumors increased levels of c-Myc and decreased expression of c-IAP2 and the central hypoxia response regulator hypoxia-inducible factor (HIF) 1α. In mice, these compounds augmented the hypoxic tumor cell death induced by cytotoxic chemotherapy, blocking angiogenesis and tumor growth. Taken together, our findings suggest that combinatorial inhibition of GSK-3β and CDK1 augment the apoptotic sensitivity of hypoxic tumors, and they offer preclinical validation of a novel and readily translatable strategy to improve cancer therapy.
Collapse
Affiliation(s)
- Patrick A. Mayes
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Nathan G. Dolloff
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA 17033
| | - Colin J. Daniel
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland OR 97239
| | - J. Judy Liu
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Lori S. Hart
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Kageaki Kuribayashi
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Joshua E. Allen
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA 17033
| | - David I.H. Jee
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Jay F. Dorsey
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Yingqiu Y. Liu
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA 17033
| | - David T. Dicker
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA 17033
| | - J. Martin Brown
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Peter S. Klein
- Departments of Medicine (Hematology/Oncology), and Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland OR 97239
| | - Wafik S. El-Deiry
- Laboratory of Molecular Oncology and Cell Cycle Regulation, Departments of Medicine (Hematology/Oncology), Genetics, and Pharmacology, the Institute for Translational Medicine and Therapeutics, and the Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State College of Medicine, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA 17033
- The American Cancer Society, Atlanta, GA 30329
| |
Collapse
|
46
|
Ko CY, Tsai MY, Tseng WF, Cheng CH, Huang CR, Wu JS, Chung HY, Hsieh CS, Sun CK, Hwang SPL, Yuh CH, Huang CJ, Pai TW, Tzou WS, Hu CH. Integration of CNS survival and differentiation by HIF2α. Cell Death Differ 2011; 18:1757-70. [PMID: 21546908 PMCID: PMC3190110 DOI: 10.1038/cdd.2011.44] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hypoxia-inducible factor (HIF) 1α and HIF2α and the inhibitor of apoptosis survivin represent prominent markers of many human cancers. They are also widely expressed in various embryonic tissues, including the central nervous system; however, little is known about their functions in embryos. Here, we show that zebrafish HIF2α protects neural progenitor cells and neural differentiation processes by upregulating the survivin orthologues birc5a and birc5b during embryogenesis. Morpholino-mediated knockdown of hif2α reduced the transcription of birc5a and birc5b, induced p53-independent apoptosis and abrogated neural cell differentiation. Depletion of birc5a and birc5b recaptured the neural development defects that were observed in the hif2α morphants. The phenotypes induced by HIF2α depletion were largely rescued by ectopic birc5a and birc5b mRNAs, indicating that Birc5a and Birc5b act downstream of HIF2α. Chromatin immunoprecipitation assay revealed that HIF2α binds to birc5a and birc5b promoters directly to modulate their transcriptions. Knockdown of hif2α, birc5a or birc5b reduced the expression of the cdk inhibitors p27/cdkn1b and p57/cdkn1c and increased ccnd1/cyclin D1 transcription in the surviving neural progenitor cells. The reduction in elavl3/HuC expression and enhanced pcna, nestin, ascl1b and sox3 expression indicate that the surviving neural progenitor cells in hif2α morphants maintain a high proliferation rate without terminally differentiating. We propose that a subset of developmental defects attributed to HIF2α depletion is due in part to the loss of survivin activity.
Collapse
Affiliation(s)
- C-Y Ko
- Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sangeetha M, Pillai MS, Philip L, Lakatta EG, Shivakumar K. NF-κB inhibition compromises cardiac fibroblast viability under hypoxia. Exp Cell Res 2011; 317:899-909. [PMID: 21211536 PMCID: PMC3216678 DOI: 10.1016/j.yexcr.2010.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 12/27/2010] [Accepted: 12/28/2010] [Indexed: 12/20/2022]
Abstract
Cardiac fibroblasts are reported to be relatively resistant to stress stimuli compared to cardiac myocytes and fibroblasts of non-cardiac origin. However, the mechanisms that facilitate their survival under conditions of stress remain unclear. We explored the possibility that NF-κB protects cardiac fibroblasts from hypoxia-induced cell death. Further, we examined the expression of the antiapoptotic cIAP-2 and Bcl-2 in hypoxic cardiac fibroblasts, and their possible regulation by NF-κB. Phase contrast microscopy and propidium iodide staining revealed that cardiac fibroblasts are more resistant than pulmonary fibroblasts to hypoxia. Electrophoretic Mobility Shift Assay showed that hypoxia activates NF-κB in cardiac fibroblasts. Supershift assay indicated that the active NF-κB complex is a p65/p50 heterodimer. An I-κB-super-repressor was constructed that prevented NF-κB activation and compromised cell viability under hypoxic but not normoxic conditions. Similar results were obtained with Bay 11-7085, an inhibitor of NF-κB. Western blot analysis showed constitutive levels of Bcl-2 and hypoxic induction of cIAP-2 in these cells. NF-κB inhibition reduced cIAP-2 but not Bcl-2 levels in hypoxic cardiac fibroblasts. The results show for the first time that NF-κB is an important effector of survival in cardiac fibroblasts under hypoxic stress and that regulation of cIAP-2 expression may contribute to its pro-survival role.
Collapse
Affiliation(s)
- M Sangeetha
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum 695011, India
| | - Malini S Pillai
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum 695011, India
| | - Linda Philip
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum 695011, India
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Gerontology Research Center, NIA/NIH, Baltimore 21224, Maryland, USA
| | - K Shivakumar
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum 695011, India
| |
Collapse
|
48
|
Tak E, Lee S, Lee J, Rashid MA, Kim YW, Park JH, Park WS, Shokat KM, Ha J, Kim SS. Human carbonyl reductase 1 upregulated by hypoxia renders resistance to apoptosis in hepatocellular carcinoma cells. J Hepatol 2011; 54:328-339. [PMID: 21056497 DOI: 10.1016/j.jhep.2010.06.045] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 06/21/2010] [Accepted: 06/22/2010] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Human carbonyl reductase1 (CBR1) has been reported to protect cells against lipid peroxidation. Since human hepatocellular carcinoma (HCC) cells are under oxidative stress in hypoxic conditions, we tested if CBR1 is upregulated by hypoxia inducible factor (HIF)-1α, helps tumor growth under hypoxia, and renders chemoresistance to cisplatin and doxorubicin in HCC. METHODS Luciferase, EMSA, and chromatin immunoprecipitation (ChIP) assays were performed to analyze whether HIF-1α transactivates CBR1 promoter. CBR1 overexpression, siRNA, and inhibitors were used to study the role of CBR1 in tumor survival under hypoxia and chemoresistance to cisplatin and doxorubicin in HCC. FACS and Western blot analysis for oxidative stress markers were performed to measure ROS. Immunohistochemistry (IHC) was performed to analyze CBR1 expression in 78 cases of HCC and 123 cases of colon cancer tissues. RESULTS The CBR1 promoter was activated by HIF-1α. CBR1 overexpression enhanced cell survival by decreasing oxidative stress under hypoxia, cisplatin, and doxorubicin treatment. CBR1-siRNA increased apoptosis via increasing oxidative stress. Combinational therapy of CBR1 inhibitors with cisplatin or doxorubicin enhanced cell death in HCC cells. IHC showed CBR1 overexpression in 56 (72%) out of 78 HCC and 88 (72%) out of 123 colon cancer cases. CONCLUSIONS Overexpressed CBR1 by HIF-1α plays important roles in tumor growth under hypoxia and chemoresistance to anticancer drugs. The inhibition of CBR1 by specific inhibitors enhances anticancer drug efficacy in HCC. Therefore, we concluded that CBR1 is a good molecular target for the development of anticancer drugs for HCC patients.
Collapse
Affiliation(s)
- Eunyoung Tak
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chen C, Zhou H, Wei F, Jiang L, Liu X, Liu Z, Ma Q. Increased levels of hypoxia-inducible factor-1α are associated with Bcl-xL expression, tumor apoptosis, and clinical outcome in chondrosarcoma. J Orthop Res 2011; 29:143-51. [PMID: 20661931 DOI: 10.1002/jor.21193] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypoxia-inducible factor (HIF)-1α is a key nuclear transcription factor that regulates the cellular response to hypoxia, and is important for solid tumor growth and survival. However, the underlying role of HIF-1α in human chondrosarcoma has not been well characterized. This study aims to investigate the expression patterns of HIF-1α in chondrosarcoma, and its association with clinicopathologic features, Bcl-xL expression, apoptosis index (AI), and overall survival of patients with chondrosarcoma. Our results shown that the protein levels of HIF-1α were increased, and the mRNA and protein levels of Bcl-xL were also increased in SW1353 cells under hypoxic conditions. In eight patients with chondrosarcoma, increased expression of HIF-1α and Bcl-xL was detected in chondrosarcoma tissues compared with the paired adjacent normal tissues. Of 34 archival specimens of chondrosarcomas, 20 (58.8%) showed high HIF-1α protein expression as compared to benign cartilage tumors. Increased HIF-1α expression was correlated with a higher pathological grade and MSTS stage of chondrosarcoma. Moreover, HIF-1α expression was significantly associated with Bcl-xL expression and AI. More significantly, the survival rate of patients with HIF-1α high tumors was significantly lower than that of patients with HIF-1α low tumors. These findings suggest that increased HIF-1α levels mediated up-regulation of Bcl-xL play a prominent role in evasion of apoptosis and tumor progression, and can be predictive for the prognosis in human chondrosarcoma.
Collapse
Affiliation(s)
- Changbao Chen
- Department of Orthopaedic Surgery, Peking University Third Hospital, 49 North Garden Road, Beijing 100191, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Gammella E, Cairo G, Tacchini L. Adenosine A(2)A receptor but not HIF-1 mediates Tyrosine hydroxylase induction in hypoxic PC12 cells. J Neurosci Res 2010; 88:2007-16. [PMID: 20143408 DOI: 10.1002/jnr.22366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tyrosine hydroxylase (TH) is the rate-limiting enzyme in the biosynthesis of catecholamines released by oxygen-sensitive cells in response to hypoxic conditions. Adenosine is released in response to hypoxia in the central nervous system and CGS21680, an adenosine A(2)A receptor agonist, induces TH transcription. As we have previously demonstrated the A(2)A receptor-mediated induction of HIF-1 in macrophages and hepatocytes, we investigated the involvement of HIF-1 in the adenosine-mediated activation of TH expression. Exposure to adenosine or CGS21680 increased TH mRNA and protein levels in PC12 cells. Transcription of a reporter gene under the control of the wild type rat TH promoter was induced 3.5-fold in CGS21680-treated cells, but neither the mutation of the hypoxia responsive element in the TH promoter nor the co-transfection of a dominant negative of the HIF-1 beta subunit prevented the increase in transcription; furthermore, CGS21680 increased CREB binding activity but did not induce HIF-1 DNA binding activity or protein levels. To investigate whether HIF-1 was involved in the hypoxia-mediated induction of TH, PC12 cells were exposed to hypoxia in the presence of the A(2)A receptor antagonist ZM241385, which prevented hypoxia-dependent TH induction despite HIF-1 activation; in line with this finding, the inhibition of HIF-1 did not abolish TH induction in hypoxic PC12 cells. These results indicate that, under hypoxic conditions, TH (a key factor in systemic adaptation to reduced oxygen availability) is not regulated by HIF-1, the primary modulator of the response to hypoxia, but by the adenosine A(2)A receptor-mediated signalling pathway.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Human Morphology and Biomedical Sciences, Città Studi, Università degli Studi di Milano, Milan, Italy
| | | | | |
Collapse
|