1
|
Zhang Z, Yu X, Wen L, Wang J, Li Z, Zhang Y, Cheng J, Kan R, Zhang W, Shen Y, Yuan S, Zhao L. USP9X integrates TGF-β and hypoxia signalings to promote ovarian cancer chemoresistance via HIF-2α-maintained stemness. Cell Death Dis 2025; 16:312. [PMID: 40246814 PMCID: PMC12006517 DOI: 10.1038/s41419-025-07646-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Widespread intraperitoneal metastases and chemoresistance render ovarian cancer the leading cause of gynecological malignancy-related deaths, wherein TGF-β signaling plays the pivotal role by promoting cancer stem cells (CSCs) activity. The activation mechanism and key protumorigeneic events downstream of TGF-β signaling remain incompletely understood. Here, we identify hypoxic tumor microenvironment as an initiator of TGF-β signaling to promote HIF-2α positive CSC-mediated chemoresistance in high-grade serous ovarian cancer (HGSOC). Mechanistically, deubiquitinase USP9X, as a TGF-β downstream effector, stabilizes HIF-2ɑ in a hydroxylation- and ubiquitylation-dependent manner, thus promoting stemness reprogramming. Hypoxia and TGF-β signals converge on USP9X-HIF-2ɑ axis via multi-level regulations, which in turn facilitates Smad/HIF responses. Clinically, USP9X expression correlates with TGF-β signatures, CSCs characteristics, EMT behaviors, and chemotherapy responsiveness, along with HIF-2ɑ. Antagonizing USP9X efficiently represses tumor formation, metastasis, CSCs occurrence, while increasing chemosensitivity in orthotopic tumors, patient-derived xenograft (PDX), organoid, and chemoresistant cell models, in part via restricting TGF-β and hypoxia activities. This study deciphers the critical role of hypoxic niche in stimulating TGF-β signaling, and a downstream USP9X-HIF-2ɑ proteostatic regulatory axis in priming the HGSOC stemness, thereby provides novel targeting venues to counteract TGF-β signaling in CSCs and meliorate clinical chemoresistance.
Collapse
Affiliation(s)
- Zhenlei Zhang
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Xiujie Yu
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Department of Pathology, Tianjin Central Hospital of Gynaecology Obsterics, Tianjin, China
| | - Liqi Wen
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Jia'nan Wang
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Zhufeng Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Zhang
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Jiayu Cheng
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Ronglin Kan
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Wanting Zhang
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Yan Shen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Department of Pathology, Tianjin Central Hospital of Gynaecology Obsterics, Tianjin, China.
| | - Shukai Yuan
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China.
| | - Li Zhao
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
2
|
Zhang L, Zhou L, Feng Q, Li Q, Ge M. Mutation of Hashimoto’s Thyroiditis and Papillary Thyroid Carcinoma Related Genes and the Screening of Candidate Genes. Front Oncol 2021; 11:813802. [PMID: 34993154 PMCID: PMC8724914 DOI: 10.3389/fonc.2021.813802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 11/13/2022] Open
Abstract
Clinical studies have shown similarities in the genetic background and biological functional characteristics between Hashimoto’s thyroiditis (HT) and papillary thyroid carcinoma (PTC), and that HT may increase risks of PTC. Here, we set to determine the gene expression specificity of HT and PTC by screening related genes or co-expressed genes and exploring their genetic correlation. Referencing the Oncomine database, HT-related genes were discovered to be expressed in many different types of thyroid cancer, such as TSHR that is highly expressed in thyroid cancer. An in-depth genetic analysis and verification of 35 cancer and paracancerous tissue pairs from patients with thyroid cancer, and 35 tissues and blood cells pairs from patients with Hashimoto’s thyroiditis was conducted. Gene chip technology research showed that TSHR, BACH2, FOXE1, RNASET2, CTLA4, PTPN22, IL2RA and other HT-related genes were all expressed in PTC, in which TSHR was significantly over-expressed in PTC patients sensitive to radioactive iodine therapy, while BACH2 was significantly under-expressed in these patients. The biologically significant candidate Tag SNP highlighted from HT-related genes was screened by the high-throughput detection method. Somatic mutations in patients with HT and PTC were detected by target region capture technique, and 75 mutations were found in patients with HT and PTC. The upstream regulatory factors of the different genes shared by HT and PTC were analyzed based on Ingenuity Pathway Analysis (IPA), and it was found that HIF-1α and PD-L1 could be used as important upstream regulatory signal molecules. These results provide a basis for screening key diagnostic genes of PTC by highlighting the relationship between some HT-related genes and their polymorphisms in the pathogenesis of PTC.
Collapse
Affiliation(s)
- Lizhuo Zhang
- Department of Head and Neck Surgery, Center of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingyan Zhou
- Department of Radiology (Ultrasound), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qingqing Feng
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nano Safety & Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Qinglin Li
- Scientific Research Department, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: MingHua Ge, ; Qinglin Li,
| | - Minghua Ge
- Department of Head and Neck Surgery, Center of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- *Correspondence: MingHua Ge, ; Qinglin Li,
| |
Collapse
|
3
|
Yu J, Wang S, Shi W, Zhou W, Niu Y, Huang S, Zhang Y, Zhang A, Jia Z. Roxadustat (FG-4592) prevents Ang II hypertension by targeting angiotensin receptors and eNOS. JCI Insight 2021; 6:e133690. [PMID: 34403364 PMCID: PMC8492313 DOI: 10.1172/jci.insight.133690] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 08/11/2021] [Indexed: 11/28/2022] Open
Abstract
The prevalence of hypertension is increasing globally, while strategies for prevention and treatment of hypertension remain limited. FG-4592 (Roxadustat) is a potentially novel, orally active small-molecule hypoxia-inducible factor (HIF) stabilizer and is being used clinically to treat chronic kidney disease (CKD) anemia. In the present study, we evaluate the effects of FG-4592 on hypertension. In an angiotensin II (Ang II) hypertension model, FG-4592 abolished hypertensive responses; prevented vascular thickening, cardiac hypertrophy, and kidney injury; downregulated AGTR1 expression; and enhanced AGTR2, endothelial NO synthase (eNOS), and HIF1α protein levels in the aortas of mice. Additionally, the levels of thiobarbituric acid reactive substances (TBARs) in blood and urine were diminished by FG-4592 treatment. In vascular smooth muscle cells, FG-4592 treatment reduced angiotensin receptor type 1 (AGTR1) and increased AGTR2 levels, while preventing Ang II–induced oxidative stress. In vascular endothelial cells, FG-4592 upregulated total and phosphorylated eNOS. Moreover, FG-4592 treatment was hypotensive in L-NAME–induced hypertension. In summary, FG-4592 treatment remarkably ameliorated hypertension and organ injury, possibly through stabilizing HIF1α and subsequently targeting eNOS, AGTR1, AGTR2, and oxidative stress. Therefore, in addition to its role in treating CKD anemia, FG-4592 could be explored as a treatment for hypertension associated with high renin angiotensin system (RAS) activity or eNOS defects.
Collapse
Affiliation(s)
- Jing Yu
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Shuqin Wang
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Wei Shi
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Yujia Niu
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Shuai Y, Duan Y, Zhou M, Yue K, Liu D, Fang Y, Wang Y, Wu Y, Zhang Z, Wang X. Development and Validation of a Nomogram based on cell growth-related Biomarkers for Oral Squamous Cell Carcinoma. J Cancer 2021; 12:5153-5163. [PMID: 34335932 PMCID: PMC8317514 DOI: 10.7150/jca.54475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 05/25/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose: We aimed to develop a prognostic nomogram based on immunohistochemistry (IHC) biomarkers of patients with oral squamous cell carcinoma (OSCC). Methods: A total of 294 patients were enrolled in the study. The least absolute shrinkage and selection operator (LASSO) Cox regression model was performed to develop a combined IHC score (IHCs) classifier. Results: Five biomarkers, specifically c-Met, Vimentin, HIF-2α, VEGF-c, and Bcl-2 were extracted. Then, an IHCs classifier was developed, and patients were stratified into high- and low-IHCs groups. In the training cohort, the 5-year overall survival (OS) was 62.1% in low-IHCs group and 28.2% in high-IHCs group (P<0.001). The 5-year OS was 68.6% for the low-IHCs group and 28.4% for the high-IHCs group in the validation cohort (P<0.001). The area under the ROC curve (AUROC) of the combination of the IHCs classifier and TNM stage was 0.746 (95% CI: 0.658-0.833) in the training cohort and 0.735 (95% CI: 0.651-0.818) in the validation cohort, respectively. Conclusions: The nomogram could effectively predict the prognosis for patients with OSCC and may be employed as a potential tool to guide the individual decision-making process.
Collapse
Affiliation(s)
- Yanjie Shuai
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Yuansheng Duan
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Mengqian Zhou
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Kai Yue
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Dandan Liu
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Yan Fang
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Yuxuan Wang
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Yansheng Wu
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Ze Zhang
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Xudong Wang
- Department of Maxillofacial & E.N.T oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| |
Collapse
|
5
|
van Vliet T, Casciaro F, Demaria M. To breathe or not to breathe: Understanding how oxygen sensing contributes to age-related phenotypes. Ageing Res Rev 2021; 67:101267. [PMID: 33556549 DOI: 10.1016/j.arr.2021.101267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
Aging is characterized by a progressive loss of tissue integrity and functionality due to disrupted homeostasis. Molecular oxygen is pivotal to maintain tissue functions, and aerobic species have evolved a sophisticated sensing system to ensure proper oxygen supply and demand. It is not surprising that aberrations in oxygen and oxygen-associated pathways subvert health and promote different aspects of aging. In this review, we discuss emerging findings on how oxygen-sensing mechanisms regulate different cellular and molecular processes during normal physiology, and how dysregulation of oxygen availability lead to disease and aging. We describe various clinical manifestations associated with deregulation of oxygen balance, and how oxygen-modulating therapies and natural oxygen oscillations influence longevity. We conclude by discussing how a better understanding of oxygen-related mechanisms that orchestrate aging processes may lead to the development of new therapeutic strategies to extend healthy aging.
Collapse
|
6
|
Hypoxia as a Driving Force of Pluripotent Stem Cell Reprogramming and Differentiation to Endothelial Cells. Biomolecules 2020; 10:biom10121614. [PMID: 33260307 PMCID: PMC7759989 DOI: 10.3390/biom10121614] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Inadequate supply of oxygen (O2) is a hallmark of many diseases, in particular those related to the cardiovascular system. On the other hand, tissue hypoxia is an important factor regulating (normal) embryogenesis and differentiation of stem cells at the early stages of embryonic development. In culture, hypoxic conditions may facilitate the derivation of embryonic stem cells (ESCs) and the generation of induced pluripotent stem cells (iPSCs), which may serve as a valuable tool for disease modeling. Endothelial cells (ECs), multifunctional components of vascular structures, may be obtained from iPSCs and subsequently used in various (hypoxia-related) disease models to investigate vascular dysfunctions. Although iPSC-ECs demonstrated functionality in vitro and in vivo, ongoing studies are conducted to increase the efficiency of differentiation and to establish the most productive protocols for the application of patient-derived cells in clinics. In this review, we highlight recent discoveries on the role of hypoxia in the derivation of ESCs and the generation of iPSCs. We also summarize the existing protocols of hypoxia-driven differentiation of iPSCs toward ECs and discuss their possible applications in disease modeling and treatment of hypoxia-related disorders.
Collapse
|
7
|
Bacigalupa ZA, Rathmell WK. Beyond glycolysis: Hypoxia signaling as a master regulator of alternative metabolic pathways and the implications in clear cell renal cell carcinoma. Cancer Lett 2020; 489:19-28. [PMID: 32512023 PMCID: PMC7429250 DOI: 10.1016/j.canlet.2020.05.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
The relationship between kidney cancer, specifically clear cell renal cell carcinoma (ccRCC), and the hypoxia signaling program has been extensively characterized. Its underlying role as the primary driver of the disease has led to the development of the most effective targeted therapies to date. Cellular responses to hypoxia or mutations affecting the von Hippel-Lindau (VHL) tumor suppressor gene stabilize the hypoxia inducible factor (HIF) transcription factors which then orchestrate elaborate downstream signaling events resulting in adaptations to key biological processes, such as reprogramming metabolism. The direct link of hypoxia signaling to glucose uptake and glycolysis has long been appreciated; however, the HIF family of proteins directly regulate many downstream targets, including other transcription factors with their own extensive networks. In this review, we will summarize our current understanding of how hypoxia signaling regulates other metabolic pathways and how this contributes to the development and progression of clear cell renal cell carcinomas.
Collapse
Affiliation(s)
- Zachary A Bacigalupa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
8
|
Wagner F, Holzapfel BM, McGovern JA, Shafiee A, Baldwin JG, Martine LC, Lahr CA, Wunner FM, Friis T, Bas O, Boxberg M, Prodinger PM, Shokoohmand A, Moi D, Mazzieri R, Loessner D, Hutmacher DW. Humanization of bone and bone marrow in an orthotopic site reveals new potential therapeutic targets in osteosarcoma. Biomaterials 2018; 171:230-246. [PMID: 29705656 DOI: 10.1016/j.biomaterials.2018.04.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Existing preclinical murine models often fail to predict effects of anti-cancer drugs. In order to minimize interspecies-differences between murine hosts and human bone tumors of in vivo xenograft platforms, we tissue-engineered a novel orthotopic humanized bone model. METHODS Orthotopic humanized tissue engineered bone constructs (ohTEBC) were fabricated by 3D printing of medical-grade polycaprolactone scaffolds, which were seeded with human osteoblasts and embedded within polyethylene glycol-based hydrogels containing human umbilical vein endothelial cells (HUVECs). Constructs were then implanted at the femur of NOD-scid and NSG mice. NSG mice were then bone marrow transplanted with human CD34 + cells. Human osteosarcoma (OS) growth was induced within the ohTEBCs by direct injection of Luc-SAOS-2 cells. Tissues were harvested for bone matrix and marrow morphology analysis as well as tumor biology investigations. Tumor marker expression was analyzed in the humanized OS and correlated with the expression in 68 OS patients utilizing tissue micro arrays (TMA). RESULTS After harvesting the femurs micro computed tomography and immunohistochemical staining showed an organ, which had all features of human bone. Around the original mouse femur new bone trabeculae have formed surrounded by a bone cortex. Staining for human specific (hs) collagen type-I (hs Col-I) showed human extracellular bone matrix production. The presence of nuclei staining positive for human nuclear mitotic apparatus protein 1 (hs NuMa) proved the osteocytes residing within the bone matrix were of human origin. Flow cytometry verified the presence of human hematopoietic cells. After injection of Luc-SAOS-2 cells a primary tumor and lung metastasis developed. After euthanization histological analysis showed pathognomic features of osteoblastic OS. Furthermore, the tumor utilized the previously implanted HUVECS for angiogenesis. Tumor marker expression was similar to human patients. Moreover, the recently discovered musculoskeletal gene C12orf29 was expressed in the most common subtypes of OS patient samples. CONCLUSION OhTEBCs represent a suitable orthotopic microenvironment for humanized OS growth and offers a new translational direction, as the femur is the most common location of OS. The newly developed and validated preclinical model allows controlled and predictive marker studies of primary bone tumors and other bone malignancies.
Collapse
Affiliation(s)
- Ferdinand Wagner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstraße 4, 80337 Munich, Germany; Department of Orthopedics for the University of Regensburg, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, 93077 Bad Abbach, Germany
| | - Boris M Holzapfel
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany
| | - Jacqui A McGovern
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Abbas Shafiee
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Jeremy G Baldwin
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Laure C Martine
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Christoph A Lahr
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Felix M Wunner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Thor Friis
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Onur Bas
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Melanie Boxberg
- Institute of Pathology, Klinikum Rechts der Isar, Technical University Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Peter M Prodinger
- Department of Orthopedic Surgery, Klinikum Rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
| | - Ali Shokoohmand
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia
| | - Davide Moi
- The University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Roberta Mazzieri
- The University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Daniela Loessner
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD 4059, Brisbane, Australia; George W Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive Northwest, Atlanta, GA 30332, USA; Institute for Advanced Study, Technical University Munich, Lichtenbergstraße 2a, 85748 Garching, Munich, Germany.
| |
Collapse
|
9
|
Zhang B, Ye H, Yang A. Mathematical modelling of interacting mechanisms for hypoxia mediated cell cycle commitment for mesenchymal stromal cells. BMC SYSTEMS BIOLOGY 2018; 12:35. [PMID: 29606139 PMCID: PMC5879778 DOI: 10.1186/s12918-018-0560-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Background Existing experimental data have shown hypoxia to be an important factor affecting the proliferation of mesenchymal stromal cells (MSCs), but the contrasting observations made at various hypoxic levels raise the questions of whether hypoxia accelerates proliferation, and how. On the other hand, in order to meet the increasing demand of MSCs, an optimised bioreactor control strategy is needed to enhance in vitro production. Results A comprehensive, single-cell mathematical model has been constructed in this work, which combines cellular oxygen sensing with hypoxia-mediated cell cycle progression to predict cell cycle commitment as a proxy to proliferation rate. With oxygen levels defined for in vitro cell culture, the model predicts enhanced proliferation under intermediate (2–8%) and mild (8–15%) hypoxia and cell quiescence under severe (< 2%) hypoxia. Global sensitivity analysis and quasi-Monte Carlo simulation revealed that within a certain range (+/− 100%), model parameters affect (with varying significance) the minimum commitment time, but the existence of a range of optimal oxygen tension could be preserved with the hypothesized effects of Hif2α and reactive oxygen species (ROS). It appears that Hif2α counteracts Hif1α and ROS-mediated protein deactivation under intermediate hypoxia and normoxia (20%), respectively, to regulate the response of cell cycle commitment to oxygen tension. Conclusion Overall, this modelling study offered an integrative framework to capture several interacting mechanisms and allowed in silico analysis of their individual and collective roles in shaping the hypoxia-mediated commitment to cell cycle. The model offers a starting point to the establishment of a suitable mechanism that can satisfactorily explain the different existing experimental observations from different studies, and warrants future extension and dedicated experimental validation to eventually support bioreactor optimisation. Electronic supplementary material The online version of this article (10.1186/s12918-018-0560-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Engineering Science, University of Oxford, Oxford, UK.,Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Martínez-Poveda B, Quesada AR, Medina MÁ. Pleiotropic Role of Puupehenones in Biomedical Research. Mar Drugs 2017; 15:md15100325. [PMID: 29065486 PMCID: PMC5666431 DOI: 10.3390/md15100325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/27/2017] [Accepted: 10/16/2017] [Indexed: 12/11/2022] Open
Abstract
Marine sponges represent a vast source of metabolites with very interesting potential biomedical applications. Puupehenones are sesquiterpene quinones isolated from sponges of the orders Verongida and Dictyoceratida. This family of chemical compounds is composed of a high number of metabolites, including puupehenone, the most characteristic compound of the family. Chemical synthesis of puupehenone has been reached by different routes, and the special chemical reactivity of this molecule has allowed the synthesis of many puupehenone-derived compounds. The biological activities of puupehenones are very diverse, including antiangiogenic, antitumoral, antioxidant, antimicrobial, immunomodulatory and antiatherosclerotic effects. Despite the very important roles described for puupehenones concerning different pathologies, the exact mechanism of action of these compounds and the putative therapeutic effects in vivo remain to be elucidated. This review offers an updated and global view about the biology of puupehenones and their therapeutic possibilities in human diseases such as cancer.
Collapse
Affiliation(s)
- Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, and IBIMA; E-29071 Málaga, Spain.
| | - Ana R Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, and IBIMA; E-29071 Málaga, Spain.
- Unidad 741 de CIBER "de Enfermedades Raras", E-29071 Málaga, Spain.
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, and IBIMA; E-29071 Málaga, Spain.
- Unidad 741 de CIBER "de Enfermedades Raras", E-29071 Málaga, Spain.
| |
Collapse
|
11
|
Martínez-Sáez O, Gajate Borau P, Alonso-Gordoa T, Molina-Cerrillo J, Grande E. Targeting HIF-2 α in clear cell renal cell carcinoma: A promising therapeutic strategy. Crit Rev Oncol Hematol 2017; 111:117-123. [PMID: 28259286 DOI: 10.1016/j.critrevonc.2017.01.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/22/2017] [Indexed: 12/24/2022] Open
Abstract
The loss of the Von Hippel-Lindau tumor suppressor (VHL) is a key oncogenic event in the vast majority of patients with clear cell renal cell carcinoma (ccRCC). With the loss of the VHL protein (pVHL) function, the hypoxia inducible factor α (HIF-α) accumulates inside the tumor cell and dimerizes with HIF-β. The HIF-α/HIF-β complex transcriptionally activates hundreds of genes promoting the adaptation to hypoxia that is implicated in tumor development. There is growing evidence showing that HIF-2α subunit has a central role in ccRCC over HIF-1α. Thus, efforts have been made to specifically target this pathway. PT2385 and PT2399 are first-in-class, orally available, small molecule inhibitors of HIF-2 that selectively disrupt the heterodimerization of HIF-2α with HIF-1β. Preclinical and clinical data indicate that these new molecules are effective in blocking cancer cell growth, proliferation, and tumor angiogenesis characteristic in ccRCC. Treatment with HIF-2α specific antagonists, either alone or in combination with immunotherapy or other antiangiogenic agents have the potential to transform the therapeutic landscape in this tumor in the future. Herein, we summarize the molecular background behind the use of HIF-2α inhibitors in ccRCC and give an overview of the development of new agents in this setting.
Collapse
Affiliation(s)
- Olga Martínez-Sáez
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain.
| | - Pablo Gajate Borau
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Enrique Grande
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| |
Collapse
|
12
|
Myszczyszyn A, Czarnecka AM, Matak D, Szymanski L, Lian F, Kornakiewicz A, Bartnik E, Kukwa W, Kieda C, Szczylik C. The Role of Hypoxia and Cancer Stem Cells in Renal Cell Carcinoma Pathogenesis. Stem Cell Rev Rep 2016. [PMID: 26210994 PMCID: PMC4653234 DOI: 10.1007/s12015-015-9611-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cancer stem cell (CSC) model has recently been approached also in renal cell carcinoma (RCC). A few populations of putative renal tumor-initiating cells (TICs) were identified, but they are indifferently understood; however, the first and most thoroughly investigated are CD105-positive CSCs. The article presents a detailed comparison of all renal CSC-like populations identified by now as well as their presumable origin. Hypoxic activation of hypoxia-inducible factors (HIFs) contributes to tumor aggressiveness by multiple molecular pathways, including the governance of immature stem cell-like phenotype and related epithelial-to-mesenchymal transition (EMT)/de-differentiation, and, as a result, poor prognosis. Due to intrinsic von Hippel-Lindau protein (pVHL) loss of function, clear-cell RCC (ccRCC) develops unique pathological intra-cellular pseudo-hypoxic phenotype with a constant HIF activation, regardless of oxygen level. Despite satisfactory evidence concerning pseudo-hypoxia importance in RCC biology, its influence on putative renal CSC-like largely remains unknown. Thus, the article discusses a current knowledge of HIF-1α/2α signaling pathways in the promotion of undifferentiated tumor phenotype in general, including some experimental findings specific for pseudo-hypoxic ccRCC, mostly dependent from HIF-2α oncogenic functions. Existing gaps in understanding both putative renal CSCs and their potential connection with hypoxia need to be filled in order to propose breakthrough strategies for RCC treatment.
Collapse
Affiliation(s)
- Adam Myszczyszyn
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| | - Damian Matak
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Lukasz Szymanski
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Fei Lian
- Emory School of Medicine, Atlanta, GA, USA
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of General Surgery and Transplantology, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Wojciech Kukwa
- Department of Otolaryngology, Czerniakowski Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| |
Collapse
|
13
|
Lee KE, Spata M, Bayne LJ, Buza EL, Durham AC, Allman D, Vonderheide RH, Simon MC. Hif1a Deletion Reveals Pro-Neoplastic Function of B Cells in Pancreatic Neoplasia. Cancer Discov 2016; 6:256-69. [PMID: 26715642 PMCID: PMC4783189 DOI: 10.1158/2159-8290.cd-15-0822] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/22/2015] [Indexed: 12/25/2022]
Abstract
UNLABELLED Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths worldwide, with an exceedingly low 5-year survival rate. PDAC tumors are characterized by an extensive desmoplastic stromal response and hypovascularity, suggesting that tumor hypoxia could regulate PDAC initiation and/or progression. Using a well-defined, autochthonous Kras(G12D)-driven murine model, as well as human tumors, we demonstrate that hypoxia and stabilization of hypoxia-inducible factor 1α (HIF1α), a principal mediator of hypoxic adaptation, emerge early during preinvasive stages of PDAC. Surprisingly, pancreas-specific Hif1a deletion drastically accelerated Kras(G12D)-driven pancreatic neoplasia and was accompanied by significant increases in intrapancreatic B lymphocytes, featuring prominent influx of a rare "B1b" B-cell subtype. Finally, treatment of HIF1α-deficient mice with B cell-depleting αCD20 monoclonal antibodies inhibited progression of pancreatic intraepithelial neoplasia (PanIN). Our data reveal a previously unrecognized role for B cells in promoting pancreatic tumorigenesis and implicate HIF1α as a critical regulator of PDAC development. SIGNIFICANCE We show here that pancreas-specific Hif1a deletion promotes PDAC initiation, coincident with increased intrapancreatic accumulation of B cells, and that B-cell depletion suppresses pancreatic tumorigenesis. We therefore demonstrate a protective role for HIF1α in pancreatic cancer initiation and uncover a previously unrecognized function of B cells.
Collapse
MESH Headings
- Animals
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Carcinoma in Situ
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Chemokine CXCL13/metabolism
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/immunology
- Disease Models, Animal
- Gene Deletion
- Genes, ras
- Humans
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/deficiency
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Mice, Knockout
- Neoplasm Invasiveness
- Neoplasm Staging
- Pancreatic Neoplasms/etiology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
Collapse
Affiliation(s)
- Kyoung Eun Lee
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Spata
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lauren J Bayne
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth L Buza
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy C Durham
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Allman
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania. Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania. Howard Hughes Medical Institute, Chevy Chase, Maryland.
| |
Collapse
|
14
|
Qian J, Wenguang X, Zhiyong W, Yuntao Z, Wei H. Hypoxia inducible factor: a potential prognostic biomarker in oral squamous cell carcinoma. Tumour Biol 2016; 37:10815-20. [DOI: 10.1007/s13277-016-4976-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/04/2016] [Indexed: 12/18/2022] Open
|
15
|
Liu YM, Ying SP, Huang YR, Pan Y, Chen WJ, Ni LQ, Xu JY, Shen QY, Liang Y. Expression of HIF-1α and HIF-2α correlates to biological and clinical significance in papillary thyroid carcinoma. World J Surg Oncol 2016; 14:30. [PMID: 26846782 PMCID: PMC4743326 DOI: 10.1186/s12957-016-0785-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/27/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The aim of this study was to detect the expression of hypoxia-inducible factor (HIF)-1α and HIF-2α in papillary thyroid carcinoma (PTC) compared with normal thyroid tissues. METHODS The mRNA levels and protein levels of HIF-1α and HIF-2α were detected by real-time PCR and Western blot separately in 30 pairs of PTCs and normal thyroid cases. The protein levels were also detected by immunohistochemistry (IHC) using 92 samples of PTC group and 46 normal samples as control group for analyzing the biological and clinical significance of the expression of HIF-1α/HIF-2α. RESULTS Real-time PCR results showed the mRNA level of HIF-1α and HIF-2α were significantly higher in PTC than normal group (P<0.001). Also, significantly higher positive rates (73%/65%) of HIF-1α and HIF-2α were observed in PTC compared with the control group (27%/35%) by IHC (P<0.01); the consistent results were gotten with Western blot. Although we did not find a significant correlation between the expression of HIF-1α and HIF-2α with gender, age, calcification, or Hashimoto's disease in the present study (P>0.05), both of their expressions were correlated to lymph node metastasis (P<0.05), capsular invasion (P<0.05), and TNM stage (P<0.05). CONCLUSIONS Overexpression of HIF-1α and HIF-2α are associated with the carcinogenesis of PTC, served as potential biomarkers of PTC.
Collapse
Affiliation(s)
- Yan-Mei Liu
- Department of Radiation Oncology, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
| | - Shen-Peng Ying
- Department of Radiation Oncology, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
| | | | - Yin Pan
- Department of Breast and Thyroid Surgery, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
| | - Wei-Jun Chen
- Department of Radiation Oncology, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
| | - Ling-Qin Ni
- Department of Radiation Oncology, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
| | - Jin-Ye Xu
- Department of Breast and Thyroid Surgery, Taizhou Municipal Hospital, Taizhou, 318000, Zhejiang, China.
| | - Qin-Yan Shen
- Department of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yong Liang
- Taizhou University, Taizhou, 318000, Zhejiang, China. .,Department of The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
16
|
A direct plasma assay of circulating microRNA-210 of hypoxia can identify early systemic metastasis recurrence in melanoma patients. Oncotarget 2016; 6:7053-64. [PMID: 25749524 PMCID: PMC4466669 DOI: 10.18632/oncotarget.3142] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/10/2015] [Indexed: 02/06/2023] Open
Abstract
Circulating cell-free(cf) microRNAs (miRNAs) have been reported to exist in plasma. MicroRNA-210(miR-210) is known to play important roles in the tumor hypoxic state. We hypothesized that the expression levels of cf-miR-210 in plasma would predict early clinical recurrence in melanoma patients. A direct miRNA assay on plasma (RT-qPCR-DP) was developed to improve cf-miRNA assay logistics, eliminate RNA extraction, and reduce specimen amount required. RNA was extracted from formalin-fixed paraffin-embedded (FFPE) melanoma tissues (n = 108) and assessed by RT-qPCR. Plasma (10 μl; n = 264) was procured from AJCC Stage III/IV patients in phase III clinical trials. A RT-qPCR-DP was performed to detect cf-miR-210. MiR-210 was significantly higher in metastatic tumors compared to primary tumors. Cf-miR-210 was significantly higher in melanoma patients versus healthy donor controls. In serial bloods within individual patients, cf-miR-210 < 3 months prior to disease recurrence significantly increased compared to baseline levels (p = 0.012). ROC curve analysis demonstrated that patients with elevated cf-miR-210 were more likely to have disease recurrence. Moreover, cf-miR-210 increase significantly correlated with poorer prognosis (p < 0.001). Lactate dehydrogenase (LDH) level was also assessed within patients, and the AIC values for proportional hazards regression models of cf-miR-210(120.01) and LDH (122.91) demonstrated that cf-miR-210 is a better recurrence indicator. We concluded enhanced cf-miR-210 provides identification of early systemic melanoma recurrence.
Collapse
|
17
|
Differential effects of HIF-α isoforms on apoptosis in renal carcinoma cell lines. Cancer Cell Int 2015; 15:23. [PMID: 25729330 PMCID: PMC4342814 DOI: 10.1186/s12935-015-0175-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/10/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Germline mutations in the von Hippel-Lindau (VHL) tumor suppressor gene predispose individuals to clear cell renal carcinomas, hemangioblastomas, and pheochromocytomas. The VHL gene product forms an ubiquitin E3 ligase complex, with regulation of hypoxia-inducible factor alpha (HIF-α) as its best known function. Lack of VHL expression has been shown previously to sensitize renal cells to apoptosis caused by certain cellular stresses. In this report, the role of HIF-α in apoptosis was investigated using two parent VHL-null renal carcinoma cell lines. METHODS 786-O and RCC10 renal carcinoma cell lines with manipulated levels of VHL, HIF-1α, or HIF-2α were subjected to cellular stresses and analyzed by western blotting for the abundance of apoptotic markers. RESULTS Cell lines expressing mutant VHL proteins that were unable to regulate HIF-α had increased levels of apoptosis when irradiated with ultraviolet (UV) light. The influences of the two major isoforms of HIF-α, HIF-1α and HIF-2α, on apoptosis, were compared by creating cell lines in which levels of each isoform were modulated via short hairpin RNA interference. In UV-irradiated cells, HIF-2α expression was determined to promote apoptosis, whereas HIF-1α was anti-apoptotic. In cells deprived of either glucose or serum, HIF-1α expression was generally anti-apoptotic, while HIF-2α expression was observed to either promote apoptosis or have less of an influence on apoptosis, depending on the cell line used. CONCLUSIONS HIF-1α and HIF-2α exerted distinct effects in each of the conditions tested, with expression of HIF-1α largely blocking apoptosis and HIF-2α generally promoting apoptosis. These results reinforce that HIF-1α and HIF-2α have distinct biological roles and that their relative expression levels may influence some therapeutic interventions that are dependent on apoptosis.
Collapse
|
18
|
The role of hypoxia-inducible factor-2 in digestive system cancers. Cell Death Dis 2015; 6:e1600. [PMID: 25590810 PMCID: PMC4669763 DOI: 10.1038/cddis.2014.565] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 01/24/2023]
Abstract
Hypoxia is an all but ubiquitous phenomenon in cancers. Two known hypoxia-inducible factors (HIFs), HIF-1α and HIF-2α, primarily mediate the transcriptional response to hypoxia. Despite the high homology between HIF-1α and HIF-2α, emerging evidence suggests differences between both molecules in terms of transcriptional targets as well as impact on multiple physiological pathways and tumorigenesis. To date, much progress has been made toward understanding the roles of HIF-2α in digestive system cancers. Indeed, HIF-2α has been shown to regulate multiple aspects of digestive system cancers, including cell proliferation, angiogenesis and apoptosis, metabolism, metastasis and resistance to chemotherapy. These findings make HIF-2α a critical regulator of this malignant phenotype. Here we summarize the function of HIF-2 during cancer development as well as its contribution to tumorigenesis in digestive system malignancies.
Collapse
|
19
|
Zhao X, Li F, Li Y, Wang H, Ren H, Chen J, Nie G, Hao J. Co-delivery of HIF1α siRNA and gemcitabine via biocompatible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials 2015; 46:13-25. [PMID: 25678112 DOI: 10.1016/j.biomaterials.2014.12.028] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/28/2014] [Accepted: 12/20/2014] [Indexed: 12/17/2022]
Abstract
Hypoxia-inducible factor 1α (HIF1α) has emerged as a promising new target for pancreatic cancer treatment over the past decade. High expression of HIF-1α increases the drug resistance of the current first line chemotherapeutic drug, gemcitabine (Gem). Here we employed biocompatible lipid-polymer hybrid nanoparticles to co-deliver HIF1α siRNA (si-HIF1α) and Gem for pancreatic cancer treatment in subcutaneous and orthotopic tumor models. The cationic ε-polylysine co-polymer (ENPs) can effectively absorb negatively charged si-HIF1α on the surface and encapsulate Gem to the hydrophilic core. Further coating of ENPs with PEGylated lipid bilayer resulted formation of LENPs, with reversed surface charge. The lipid bilayer of LENPs prevented nanoparticle aggregation and si-HIF1α degradation in serum, as well as Gem leakage. Those characteristics endow LENPs encapsulating drug prolonged lifetime in bloodstream and improved drug release via the enhanced tumor vasculature effect in tumor tissues. LENPs can co-deliver Gem and si-HIF1α (LENP-Gem-si-HIF1α) into tumor cells and effectively suppress the HIF1α expression both in vitro and in vivo. LENP-Gem-siHIF1α exhibited significant synergistic antitumor effects. Furthermore, LENP-Gem-si-HIF1α showed excellent capability to inhibit tumor metastasis in orthotopic tumor model.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Feng Li
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - He Ren
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jing Chen
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology (NCNST), 11 Beiyitiao, Zhongguancun, Beijing 100190, China.
| | - Jihui Hao
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
20
|
Yoo S, Takikawa S, Geraghty P, Argmann C, Campbell J, Lin L, Huang T, Tu Z, Feronjy R, Spira A, Schadt EE, Powell CA, Zhu J. Integrative analysis of DNA methylation and gene expression data identifies EPAS1 as a key regulator of COPD. PLoS Genet 2015; 11:e1004898. [PMID: 25569234 PMCID: PMC4287352 DOI: 10.1371/journal.pgen.1004898] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 11/17/2014] [Indexed: 01/11/2023] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a complex disease. Genetic, epigenetic, and environmental factors are known to contribute to COPD risk and disease progression. Therefore we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity. EPAS1 is distinct in comparison with other key regulators in terms of methylation profile and downstream target genes. Genes predicted to be regulated by EPAS1 were enriched for biological processes including signaling, cell communications, and system development. We confirmed that EPAS1 protein levels are lower in human COPD lung tissue compared to non-disease controls and that Epas1 gene expression is reduced in mice chronically exposed to cigarette smoke. As EPAS1 downstream genes were significantly enriched for hypoxia responsive genes in endothelial cells, we tested EPAS1 function in human endothelial cells. EPAS1 knockdown by siRNA in endothelial cells impacted genes that significantly overlapped with EPAS1 downstream genes in lung tissue including hypoxia responsive genes, and genes associated with emphysema severity. Our first integrative analysis of genome-wide DNA methylation and gene expression profiles illustrates that not only does DNA methylation play a ‘causal’ role in the molecular pathophysiology of COPD, but it can be leveraged to directly identify novel key mediators of this pathophysiology. Chronic Obstructive Pulmonary Disease (COPD) is a common lung disease. It is the fourth leading cause of death in the world and is expected to be the third by 2020. COPD is a heterogeneous and complex disease consisting of obstruction in the small airways, emphysema, and chronic bronchitis. COPD is generally caused by exposure to noxious particles or gases, most commonly from cigarette smoking. However, only 20–25% of smokers develop clinically significant airflow obstruction. Smoking is known to cause epigenetic changes in lung tissues. Thus, genetics, epigenetic, and their interaction with environmental factors play an important role in COPD pathogenesis and progression. Currently, there are no therapeutics that can reverse COPD progression. In order to identify new targets that may lead to the development of therapeutics for curing COPD, we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity.
Collapse
Affiliation(s)
- Seungyeul Yoo
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Sachiko Takikawa
- Division of Pulmonary, Critical Care and Sleep Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Patrick Geraghty
- Department of Medicine, St. Luke's Roosevelt Medical Center, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Carmen Argmann
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Joshua Campbell
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Luan Lin
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Tao Huang
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Robert Feronjy
- Department of Medicine, St. Luke's Roosevelt Medical Center, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Avrum Spira
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Eric E. Schadt
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Charles A. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Lin K, Ye P, Liu J, He F, Xu W. Endostar inhibits hypoxia-induced cell proliferation and migration via the hypoxia-inducible factor-1α/vascular endothelial growth factor pathway in vitro. Mol Med Rep 2014; 11:3780-5. [PMID: 25543905 DOI: 10.3892/mmr.2014.3131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 06/02/2014] [Indexed: 11/06/2022] Open
Abstract
Endostar, a recombinant human endostatin, is recognized as one of the most effective angiogenesis inhibitors. The angiogenesis inhibitory effects of Endostar suggest a possible beneficial role of Endostar in choroidal neovascularization (CNV), which is predominantly induced by hypoxia. In our previous study, it was reported that Endostar may inhibit the proliferation and migration of RF/6A choroid‑retinal endothelial cells. However, the inhibitory effect of Endostar on hypoxia‑induced cell proliferation and migration in RF/6A cells has not yet been elucidated. Therefore, the present study investigated the effect of Endostar on hypoxia‑induced cell proliferation and migration in RF/6A cells and the possible mechanisms underlying this effect. Under chemical hypoxia conditions, cell viability was increased to 114.9±10.1 and 123.6±9.6% in cells treated with 100 and 200 µm CoCl2, respectively, compared with the control (P<0.01). Pretreatment with 10‑100 µg/ml Endostar significantly inhibited CoCl2‑induced cell proliferation (P<0.05), and pre‑treatment with 10 µg/ml Endostar for 24, 48 and 96 h attenuated CoCl2‑promoted cell migration by 60.5, 48.3 and 39.6%, respectively, compared with the control (P<0.001). In addition, pretreatment with 10 µg/ml Endostar reversed the cell cycle arrest at S phase and the increased expression of hypoxia‑inducible factor‑1α (HIF‑1α) and vascular endothelial growth factor (VEGF) mRNA in RF/6A cells treated with 200 µM CoCl2. These data indicate that Endostar inhibited CoCl2‑induced hypoxic proliferation and migration, and limited cell cycle progression in vitro possibly through the HIF‑1α/VEGF pathway.
Collapse
Affiliation(s)
- Kana Lin
- Department of Clinical Pharmacology, The Second Affiliated Hospital (Binjiang Branch), School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310052, P.R. China
| | - Panpan Ye
- Eye Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jian Liu
- Eye Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Fengying He
- Eye Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen Xu
- Eye Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
22
|
Bordji K, Grandval A, Cuhna-Alves L, Lechapt-Zalcman E, Bernaudin M. Hypoxia-inducible factor-2α (HIF-2α), but not HIF-1α, is essential for hypoxic induction of class III β-tubulin expression in human glioblastoma cells. FEBS J 2014; 281:5220-36. [DOI: 10.1111/febs.13062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/29/2014] [Accepted: 09/18/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Karim Bordji
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Alexandra Grandval
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Leilane Cuhna-Alves
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Emmanuèle Lechapt-Zalcman
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
- CHU de Caen; Service d'Anatomie et Cytologie Pathologique; Caen France
| | - Myriam Bernaudin
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| |
Collapse
|
23
|
Wu CY, Guo XZ, Li HY. Hypoxia and Serum deprivation protected MiaPaCa-2 cells from KAI1-induced proliferation inhibition through autophagy pathway activation in solid tumors. Clin Transl Oncol 2014; 17:201-8. [PMID: 25199507 DOI: 10.1007/s12094-014-1211-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/31/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE KAI1 closely correlates with pancreatic cancer metastasis. There might be some factors that protect the cells from a proliferation inhibition by KAI1 in the solid tumors' microenvironment. Hypoxia and ischemia are the main characteristics of the microenvironment within solid tumors. Whether they affect the KAI1 inhibitory effects on cell proliferation is still unclear. METHODS MiaPaCa-2 human pancreatic cancer cells do not express KAI1 protein. However, after being infected with Ad5-KAI1, they expressed KAI1 protein. We cultured them under hypoxic and serum-free conditions to simulate the solid tumor hypoxic-ischemic microenvironment. The cells were divided into the control, hypoxic, serum-free, and hypoxic with serum-free groups. The proliferation and apoptosis were observed by CCK8 and Annexin V-FITC/PI, respectively. The green fluorescent protein-labeled light chain 3 association with autophagosome membranes was detected by confocal microscopy. The ratio of LC3-II-LC3-I expression level was detected by western blot. Pretreatment of 3-MA was used to inhibit the autophagy. We, then observed whether the hypoxic and serum-free conditions could change the effect of KAI1 on cell survival and whether the pretreatment of 3-MA could inhibit the effect of hypoxic and serum-free conditions on KAI1 function. RESULTS Hypoxia and serum-free media effectively reduced the apoptosis and proliferation inhibition caused by KAI1 and was beneficial to the cell survival. 3-MA pretreatment effectively blocked the protective effect of hypoxia and serum-free media on the cells by autophagy block. CONCLUSIONS Serum-free media and hypoxia protected the MiaPaCa-2 cells from a KAI1-induced apoptosis and proliferation inhibition via autophagy induction.
Collapse
Affiliation(s)
- C Y Wu
- Department of Gastroenterology, Shenyang General Hospital of PLA, No. 83 Wenhua Road, Shenyang, 110016, China
| | | | | |
Collapse
|
24
|
Zecchini V, Madhu B, Russell R, Pértega-Gomes N, Warren A, Gaude E, Borlido J, Stark R, Ireland-Zecchini H, Rao R, Scott H, Boren J, Massie C, Asim M, Brindle K, Griffiths J, Frezza C, Neal DE, Mills IG. Nuclear ARRB1 induces pseudohypoxia and cellular metabolism reprogramming in prostate cancer. EMBO J 2014; 33:1365-82. [PMID: 24837709 PMCID: PMC4194125 DOI: 10.15252/embj.201386874] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 04/13/2014] [Accepted: 04/17/2014] [Indexed: 12/23/2022] Open
Abstract
Tumour cells sustain their high proliferation rate through metabolic reprogramming, whereby cellular metabolism shifts from oxidative phosphorylation to aerobic glycolysis, even under normal oxygen levels. Hypoxia-inducible factor 1A (HIF1A) is a major regulator of this process, but its activation under normoxic conditions, termed pseudohypoxia, is not well documented. Here, using an integrative approach combining the first genome-wide mapping of chromatin binding for an endocytic adaptor, ARRB1, both in vitro and in vivo with gene expression profiling, we demonstrate that nuclear ARRB1 contributes to this metabolic shift in prostate cancer cells via regulation of HIF1A transcriptional activity under normoxic conditions through regulation of succinate dehydrogenase A (SDHA) and fumarate hydratase (FH) expression. ARRB1-induced pseudohypoxia may facilitate adaptation of cancer cells to growth in the harsh conditions that are frequently encountered within solid tumours. Our study is the first example of an endocytic adaptor protein regulating metabolic pathways. It implicates ARRB1 as a potential tumour promoter in prostate cancer and highlights the importance of metabolic alterations in prostate cancer.
Collapse
Affiliation(s)
- Vincent Zecchini
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Basetti Madhu
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Roslin Russell
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Nelma Pértega-Gomes
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
| | - Anne Warren
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Edoardo Gaude
- Medical Research Council Cancer Cell Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Joana Borlido
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Rory Stark
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Roheet Rao
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Helen Scott
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joan Boren
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Charlie Massie
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Mohammad Asim
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Kevin Brindle
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - John Griffiths
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Christian Frezza
- Medical Research Council Cancer Cell Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - David E Neal
- Department of CRUK, CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ian G Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership University of Oslo and Oslo University Hospital, Oslo, Norway Department of Cancer Prevention and Urology, Institute of Cancer Research and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Chi W, Gan X, Xiao W, Wang W, He S. Different evolutionary patterns of hypoxia-inducible factor α (HIF-α) isoforms in the basal branches of Actinopterygii and Sarcopterygii. FEBS Open Bio 2013; 3:479-83. [PMID: 24265980 PMCID: PMC3836196 DOI: 10.1016/j.fob.2013.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 01/23/2023] Open
Abstract
Hypoxia-inducible factor (HIF) is a crucial regulator of cellular and systemic responses to low oxygen levels. Here we firstly cloned three HIF-α isoforms from the basal branches of Osteichthyes and used computational tools to characterise the molecular change underlying the functional divergence of HIF-α isoforms in different lineages. Only the HIF-1α and HIF-2α in African lungfish and amphibians were found under positive selection. HIF-1α and -2α were less functionally divergent in basal ray-finned fish than in teleosts, and showed conserved but different transcriptional activity towards specific target genes. All three HIF-α isoforms are well preserved in basal ray-finned fish. The HIF-1α and -2α in amphibians and lungfish are positively selected. The HIF-1α and -2α are more functionally diverged in teleosts. The HIF-1α and -2α in different lineages exhibit different levels of activity.
Collapse
Affiliation(s)
- Wei Chi
- Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, 7# Donghu South Road, Wuhan, Hubei 430072, China
| | | | | | | | | |
Collapse
|
26
|
Desimone MC, Rathmell WK, Threadgill DW. Pleiotropic effects of the trichloroethylene-associated P81S VHL mutation on metabolism, apoptosis, and ATM-mediated DNA damage response. J Natl Cancer Inst 2013; 105:1355-64. [PMID: 23990666 PMCID: PMC3776265 DOI: 10.1093/jnci/djt226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The risk relevance of the P81S von Hippel-Lindau (VHL) gene hotspot mutation identified in clear cell renal cell carcinoma from individuals exposed occupationally to trichloroethylene (TCE) is not known. VHL mutations in hereditary VHL syndrome strongly correlate with phenotypic associations, but specific sporadic mutations in VHL that uniquely alter its protein function may provide a selective growth advantage for somatic cells harboring these mutations. METHODS VHL deficient (Vhl (-/-) ) mouse embryonic stem cells were generated that stably express wild-type, P81S, or R167Q human VHL protein. Under hypoxic conditions, cell lines were examined for hypoxia-inducible transcription factor family (HIF) stabilization and E3-ubiquitin ligase complex interactions. In vivo, teratomas were examined for tumor size, proliferation, apoptosis, and immunohistochemistry and subjected to gene expression analysis. Wild-type, R167Q, and P81S VHL-expressing teratomas were also exposed to 5 Gy ionizing radiation to quantify apoptotic response. Proliferation and apoptosis and teratoma growth were analyzed by either Student t test or analysis of variance with Bonferroni correction. All statistical tests were two-sided. RESULTS The P81S VHL mutation produces deregulation of HIF factors in cell culture but exhibits a growth advantage in the tumor microenvironment, in part because of suppression of apoptosis (P81S mean = 0.9%, 95% confidence interval = 0.6 to 1.2%; WT mean = 7.6%; 95% confidence interval = 6.4 to 8.8%; P < .001) coupled with sustained proliferation. Transcriptional analysis of P81S teratomas revealed the induction of metabolic pathways, antiapoptotic genes, and global suppression of key DNA damage response genes not observed in VHL wild-type or R167Q mutants. In vivo irradiation exposure showed that P81S mutant is resistant to ionizing radiation-induced apoptosis. CONCLUSIONS The TCE-associated P81S VHL mutation can initiate a unique adaptive response required for selective tumor growth through pleiotropic effects on metabolic diversification, apoptosis suppression, and alteration of the DNA damage response.
Collapse
Affiliation(s)
- Michelle C Desimone
- Affiliations of authors: Department of Genetics, North Carolina State University, Raleigh, NC (MCD, DWT); Department of Genetics (WKR) and Lineberger Cancer Center, University of North Carolina, Chapel Hill, NC (WKR, DWT)
| | | | | |
Collapse
|
27
|
McCormick RI, Blick C, Ragoussis J, Schoedel J, Mole DR, Young AC, Selby PJ, Banks RE, Harris AL. miR-210 is a target of hypoxia-inducible factors 1 and 2 in renal cancer, regulates ISCU and correlates with good prognosis. Br J Cancer 2013; 108:1133-42. [PMID: 23449350 PMCID: PMC3619073 DOI: 10.1038/bjc.2013.56] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 12/20/2012] [Accepted: 01/08/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Clear cell renal cancer frequently harbours von Hippel-Lindau (VHL) gene mutations, leading to stabilisation of the hypoxia-inducible factors (HIFs) and expression of their target genes. We investigated HIF-1 and HIF-2 in the regulation of microRNA-210 (miR-210), and its clinical relevance in renal tumours. METHODS RCC4 and 786-O renal cancer cell lines transfected with either an empty vector or functional VHL and incubated in normoxia or hypoxia were examined for miR-210 expression. Hypoxia-inducible factor siRNAs were used to examine their regulation of miR-210. Seventy-one clear cell renal tumours were sequenced for VHL mutations. Expression of miR-210, VHL, CA9, ISCU and Ki-67 were determined by immunohistochemistry and qRT-PCR. RESULTS In addition to HIF-1 regulating miR-210 in renal cancer, HIF-2 can regulate this microRNA in the absence of HIF-1. MicroRNA-210 is upregulated in renal cancer compared with normal renal cortex tissue. MicroRNA-210 correlates negatively with its gene target ISCU at the protein and mRNA level. MicroRNA-210 correlated with positive outcome variables and negatively with Ki-67. CONCLUSION We provide further evidence of miR-210 activity in vivo, and show that high miR-210 expression is associated with better clinico-pathological prognostic factors.
Collapse
Affiliation(s)
- R I McCormick
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - C Blick
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - J Ragoussis
- Genomic Research Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - J Schoedel
- Oxygen Sensing Group, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - D R Mole
- Oxygen Sensing Group, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - A C Young
- Oxygen Sensing Group, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - P J Selby
- Cancer Research UK Centre, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - R E Banks
- Cancer Research UK Centre, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - A L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| |
Collapse
|
28
|
Luan Y, Gao C, Miao Y, Li Y, Wang Z, Qiu X. Clinicopathological and prognostic significance of HIF-1α and HIF-2α expression in small cell lung cancer. Pathol Res Pract 2013; 209:184-9. [PMID: 23375698 DOI: 10.1016/j.prp.2012.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 09/24/2012] [Accepted: 10/21/2012] [Indexed: 01/16/2023]
Abstract
Previous work from our laboratory has demonstrated that urokinase plasminogen activator receptor (uPAR) may be a potential stem-like cell marker in SCLC. Hypoxia inducible factor (HIF) has been shown to transcriptionally regulate uPAR expression. Therefore, the aim of this study was to evaluate the relationship between HIF-1α/HIF-2α and uPAR expression, and to investigate the role of HIF-1α/HIF-2α in the clinical pathology and prognosis of patients with SCLC. Immunohistochemical analysis showed that HIF-1α/HIF-2α staining was mainly present in the nuclei of cancer cells. HIF-1α-positive cells were diffusely distributed in the nests of the tumor, while HIF-2α-positive cells were frequently distributed around necrotic regions. HIF-1α and HIF-2α were expressed in 22/45 (48.9%) and in 11/45 (24.4%) of SCLC patients, respectively; HIF-1α did not correlate with any of the clinicopathological parameters as evaluated in our study. In contrast, a significant association of HIF-2α with uPAR expression, tumor growth and distant metastasis (p=0.001, 0.010 and 0.008, respectively) was noted; Kaplan-Meier survival analysis demonstrated that HIF-1α and HIF-2α expressions were related to shortened overall survival (p=0.027 and 0.001, respectively). However, in multivariate analysis, only HIF-2α expression and distant metastasis were the independent prognostic indicators of SCLC (p=0.004 and 0.018, respectively). Our results suggest that HIF-2α may represent a more aggressive phenotype in SCLC. HIF-2α, in addition to HIF-1α, needs to be considered when developing drugs that target HIF pathway.
Collapse
Affiliation(s)
- Yajing Luan
- Department of Pathology, Tianjin Medical University, Tianjin, China.
| | | | | | | | | | | |
Collapse
|
29
|
Zhang B, Guo W, Yu L, Wang F, Xu Y, Liu Y, Huang C. Cobalt chloride inhibits tumor formation in osteosarcoma cells through upregulation of HIF-1α. Oncol Lett 2013; 5:911-916. [PMID: 23426939 PMCID: PMC3576193 DOI: 10.3892/ol.2013.1127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/21/2012] [Indexed: 12/04/2022] Open
Abstract
The exact effect of hypoxia on cancer development is controversial. The present study investigates the ability of osteosarcoma to form tumors in the hypoxic microenvironment induced by CoCl2. MG63 human osteosarcoma cells were cultured with different concentrations (0, 150 and 300 μM) of CoCl2 for 24 h to simulate hypoxia in vitro. The expression of hypoxia-inducible factor (HIF)-1α was analyzed by western blotting. The proliferation and drug resistance of MG63 cells were examined using the CCK-8 assay, the apoptosis rate was detected by flow cytometry, the ability to form spheroids was assessed by a sarcosphere culture system and invasiveness was determined by a vertical invasion assay. A transplantation assay was used to evaluate the ability to form tumors in vivo. Our results showed that the proliferation of MG63 cells was inhibited by treatment with CoCl2, while no effect on drug toxicity was observed. The apoptotic rate was increased in a dose-dependent manner, the ability to form sarcospheroids was suppressed, the invasiveness was inhibited and the expression of HIF-1α was upregulated following CoCl2 treatment. We also found that the ability to form tumors in vivo was inhibited. In conclusion, we provide strong evidence that CoCl2 has the ability to inhibit osteosarcoma development; the mechanism may be related to the hypoxic microenvironment and HIF-1α may be a critical regulatory factor.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan 430060, P.R. China
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhang H, Pu J, Qi T, Qi M, Yang C, Li S, Huang K, Zheng L, Tong Q. MicroRNA-145 inhibits the growth, invasion, metastasis and angiogenesis of neuroblastoma cells through targeting hypoxia-inducible factor 2 alpha. Oncogene 2012; 33:387-97. [PMID: 23222716 DOI: 10.1038/onc.2012.574] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 10/01/2012] [Accepted: 10/23/2012] [Indexed: 11/10/2022]
Abstract
Recent evidence shows that hypoxia-inducible factor 2 alpha (HIF-2α) may have critical roles in the growth and progression of neuroblastoma (NB) under non-hypoxic conditions. However, the underlying mechanisms and clinical potentials of normoxic HIF-2α expression in NB still remain largely unknown. In this study, HIF-2α immunostaining was identified in 26/42 NB tissues, which was correlated with clinicopathological features. In subtotal 20 NB cases, microRNA-145 (miR-145) was downregulated and inversely correlated with HIF-2α expression. Bioinformatics analysis revealed a putative miR-145 binding site in the 3'-untranslated region (3'-UTR) of HIF-2α messenger RNA (mRNA). Overexpression or knockdown of miR-145 responsively altered both the mRNA and protein levels of HIF-2α and its downstream genes, cyclin D1, matrix metalloproteinase 14 and vascular endothelial growth factor, in normoxically cultured NB cell lines SH-SY5Y and SK-N-SH. In a luciferase reporter system, miR-145 downregulated the luciferase activity of HIF-2α 3'-UTR, and these effects were abolished by a mutation in the putative miR-145-binding site. Overexpression of miR-145 suppressed the growth, invasion, metastasis and angiogenesis of SH-SY5Y and SK-N-SH cells in vitro and in vivo, while restoration of HIF-2α expression rescued the tumor cells from miR-145-mediated defects in these biological features. Furthermore, anti-miR-145 inhibitor rescued the HIF-2α knockdown-mediated repression on the growth, migration, invasion and angiogenesis of NB cells. These data indicate that miR-145 suppresses HIF-2α expression via the binding site in the 3'-UTR under normoxic conditions, thus inhibiting the aggressiveness and angiogenesis of NB.
Collapse
Affiliation(s)
- H Zhang
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - J Pu
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - T Qi
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - M Qi
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - C Yang
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - S Li
- Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - K Huang
- 1] Clinical Center of Human Genomic Research, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China [2] Department of Cardiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - L Zheng
- 1] Clinical Center of Human Genomic Research, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China [2] Department of Pathology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Q Tong
- 1] Department of Pediatric Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China [2] Clinical Center of Human Genomic Research, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| |
Collapse
|
31
|
McKee TC, Rabe D, Bokesch HR, Grkovic T, Whitson EL, Diyabalanage T, Van Wyk AWW, Marcum SR, Gardella RS, Gustafson KR, Linehan WM, McMahon JB, Bottaro DP. Inhibition of hypoxia inducible factor-2 transcription: isolation of active modulators from marine sponges. JOURNAL OF NATURAL PRODUCTS 2012; 75:1632-6. [PMID: 22928967 PMCID: PMC3466586 DOI: 10.1021/np300211x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Renal or kidney cancer accounts for about 3% of all cancer cases reported each year in the U.S. Molecular signatures that define the cancer, such as the loss of functional VHL, are found in both sporadic and familial cases of cancer. In clear cell renal cancer, the transcription factor HIF-2α has been shown to have a distinct role in tumorigenesis. Our laboratories developed a cell-based screen to identify modulators of HIF-2α. Screening of the NCI's Natural Product Extract Repository resulted in the identification of 10 sponge extracts, from which 12 compounds were isolated. The biological evaluation of these compounds will be discussed including evaluation of HIF-1α vs HIF-2α selectivity and the isolated compounds' effects on mRNA from several pathways regulated by HIF.
Collapse
Affiliation(s)
- Tawnya C McKee
- Molecular Targets Laboratory, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chiavarina B, Martinez-Outschoorn UE, Whitaker-Menezes D, Howell A, Tanowitz HB, Pestell RG, Sotgia F, Lisanti MP. Metabolic reprogramming and two-compartment tumor metabolism: opposing role(s) of HIF1α and HIF2α in tumor-associated fibroblasts and human breast cancer cells. Cell Cycle 2012; 11:3280-9. [PMID: 22894905 DOI: 10.4161/cc.21643] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypoxia-inducible factor (HIF) 1α and 2α are transcription factors responsible for the cellular response to hypoxia. The functional roles of HIF1α and HIF2α in cancer are distinct and vary among different tumor types. The aim of this study was to evaluate the compartment-specific role(s) of HIF1α and HIF2α in breast cancer. To this end, immortalized human fibroblasts and MDA-MB-231 breast cancer cells carrying constitutively active HIF1α or HIF2α mutants were analyzed with respect to their metabolic function(s) and ability to promote tumor growth in an in vivo setting. We observed that activation of HIF1α, but not HIF2α, in stromal cells promotes a shift toward aerobic glycolysis, with increased L-lactate production and a loss of mitochondrial activity. In a xenograft model, HIF1α-activated fibroblasts promoted the tumor growth of co-injected MDA-MB-231 cells without an increase in angiogenesis. Conversely, HIF2α-activated stromal cells did not favor tumor growth and behaved as the empty vector controls. Similarly, activation of HIF1α, but not HIF2α, in MDA-MB-231 cells promoted a shift toward aerobic glycolysis, with increased glucose uptake and L-lactate production. In contrast, HIF2α activation in cancer cells increased the expression of EGFR, Ras and cyclin D1, which are known markers of tumor growth and cell cycle progression. In a xenograft model, HIF1α activation in MDA-MB-231 cells acted as a tumor suppressor, resulting in an almost 2-fold reduction in tumor mass and volume. Interestingly, HIF2α activation in MDA-MB-231 cells induced a significant ~2-fold-increase in tumor mass and volume. Analysis of mitochondrial activity in these tumor xenografts using COX (cytochrome C oxidase) staining demonstrated elevated mitochondrial oxidative metabolism (OXPHOS) in HIF2α-tumors. We conclude that the role(s) of HIF1α and HIF2α in tumorigenesis are compartment-specific. HIF1α acts as a tumor promoter in stromal cells but as a tumor suppressor in cancer cells. Conversely, HIF2α is a tumor promoter in cancer cells. Mechanistically, HIF1α-driven aerobic glycolysis in stromal cells supports cancer cell growth via the paracrine production of nutrients (such as L-lactate) that can "feed" cancer cells. However, HIF1α-driven aerobic glycolysis in cancer cells inhibits tumor growth. Finally, HIF2α activation in cancer cells induces the expression of known pro-oncogenic molecules and promotes the mitochondrial activity of cancer cells.
Collapse
Affiliation(s)
- Barbara Chiavarina
- The Jefferson Stem Cell Biology and Regenerative Medicine Center, Department of Stem Cell Biology & Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Zeng ZJ, Johansson E, Hayashi A, Chavali PL, Akrap N, Yoshida T, Kohno K, Izumi H, Funa K. TLX controls angiogenesis through interaction with the von Hippel-Lindau protein. Biol Open 2012; 1:527-35. [PMID: 23213445 PMCID: PMC3509445 DOI: 10.1242/bio.2012893] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
TLX is known as the orphan nuclear receptor indispensable for maintaining neural stem cells in adult neurogenesis. We report here that neuroblastoma cell lines express high levels of TLX, which further increase in hypoxia to enhance the angiogenic capacity of these cells. The proangiogenetic activity of TLX appears to be induced by its direct binding to the von Hippel-Lindau protein (pVHL), which stabilizes TLX. In turn, TLX competes with hydroxylated hypoxia-inducible factor (HIF-α) for binding to pVHL, which contributes to the stabilization of HIF-2α in neuroblastoma during normoxia. Upon hypoxia, TLX increases in the nucleus where it binds in close proximity of the HIF-response element on the VEGF-promoter chromatin, and, together with HIF-2α, recruits RNA polymerase II to induce VEGF expression. Conversely, depletion of TLX by shRNA decreases the expression of HIF-2α and VEGF as well as the growth-promoting and colony-forming capacity of the neuroblastoma cell lines IMR-32 and SH-SY5Y. On the contrary, silencing HIF-2α will slightly increase TLX, suggesting that TLX acts to maintain a hypoxic environment when HIF-2α is decreasing. Our results demonstrate TLX to play a key role in controlling angiogenesis by regulating HIF-2α. TLX and pVHL might counterbalance each other in important fate decisions such as self-renewal and differentiation, as well as angiogenesis and anti-angiogenesis.
Collapse
Affiliation(s)
- Zhao-Jun Zeng
- Sahlgrenska Cancer Center, University of Gothenburg , Box 425, SE 405 30 Gothenburg , Sweden ; Molecular Biology Research Center, School of Biological Science and Technology, Central South University , Changsha 410078 , China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dai ZJ, Gao J, Ma XB, Yan K, Liu XX, Kang HF, Ji ZZ, Guan HT, Wang XJ. Up-regulation of hypoxia inducible factor-1α by cobalt chloride correlates with proliferation and apoptosis in PC-2 cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:28. [PMID: 22453051 PMCID: PMC3359273 DOI: 10.1186/1756-9966-31-28] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 03/27/2012] [Indexed: 11/25/2022]
Abstract
Background The exact mechanism of the effects of hypoxia on the proliferation and apoptosis in carcinoma cells is still conflicting. This study investigated the variation of hypoxia-inducible factor-1α(HIF-1α) expression and the apoptosis effect of hypoxia stimulated by cobalt chloride (CoCl2) in pancreatic cancer PC-2 cells. Methods PC-2 cells were cultured with different concentration (50-200 μmol/L) of CoCl2 after 24-120 hours to simulate hypoxia in vitro. The proliferation of PC-2 cells was examined by MTT assay. The cellular morphology of PC-2 cells were observed by light inverted microscope and transmission electron microscope(EM). The expression of HIF-1α on mRNA and protein level was measured by semi-quantitive RT-PCR and Western blot analysis. Apoptosis of PC-2 cells were demonstrated by flow cytometry with Annexin V-FITC/PI double staining. Results MTT assay showed that the proliferation of PC-2 cells were stimulated in the first 72 h, while after treated over 72 h, a dose- dependent inhibition of cell growth could be observed. By using transmission electron microscope, swollen chondrosomes, accumulated chromatin under the nuclear membrane and apoptosis bodies were observed. Flow cytometer(FCM) analysis showed the apoptosis rate was correlated with the dosage of CoCl2. RT-PCR and Western blot analysis indicated that hypoxia could up-regulate the expression of HIF-1α on both mRNA and protein levels. Conclusion Hypoxic microenvironment stimulated by CoCl2 could effectively induce apoptosis and influence cell proliferation in PC-2 cells, the mechanism could be related to up-expression of HIF-1α.
Collapse
Affiliation(s)
- Zhi-Jun Dai
- Department of Oncology, the Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kaelin WG. Cancer and altered metabolism: potential importance of hypoxia-inducible factor and 2-oxoglutarate-dependent dioxygenases. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2011; 76:335-45. [PMID: 22089927 DOI: 10.1101/sqb.2011.76.010975] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hypoxia-inducible factor (HIF) deregulation contributes to the Warburg effect. HIF consists of an unstable α subunit and a stable β subunit. In the presence of oxygen, HIFα becomes prolyl hydroxylated by members of the EglN (also called PHD) family, leading to its proteasomal degradation. Under hypoxic conditions, EglN activity is diminished and HIF levels rise. EglN1 is the primary HIF prolyl hydroxylase with EglN2 and EglN3 playing compensatory roles under certain conditions. EglN2 and EglN3 also appear to play HIF-independent roles in regulating cell proliferation and apoptosis, respectively. The EglNs belong to a large family of 2-oxoglutarate-dependent dioxygenases that includes the TET DNA hydroxymethylases and JmjC-containing histone demethylases. Members of this superfamily can be inhibited by endogenous metabolites, including fumarate and succinate, which accumulate in tumors that have fumarate hydratase (FH) or succinate dehydrogenase (SDH) mutations, respectively, as well as by the 2-hydroxyglutarate detected in isocitrate dehydrogenase (IDH) mutant tumors. 2-Oxoglutarate-dependent dioxygenases therefore provide a link between altered metabolism and cancer.
Collapse
Affiliation(s)
- W G Kaelin
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA.
| |
Collapse
|
36
|
Shen C, Beroukhim R, Schumacher SE, Zhou J, Chang M, Signoretti S, Kaelin WG. Genetic and functional studies implicate HIF1α as a 14q kidney cancer suppressor gene. Cancer Discov 2011; 1:222-35. [PMID: 22037472 PMCID: PMC3202343 DOI: 10.1158/2159-8290.cd-11-0098] [Citation(s) in RCA: 347] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
UNLABELLED Kidney cancers often delete chromosome 3p, spanning the VHL tumor suppressor gene, and chromosome 14q, which presumably harbors ≥ 1 tumor suppressor genes. pVHL inhibits the hypoxia-inducible transcription factor (HIF), and HIF2α is a kidney cancer oncoprotein. In this article, we identify focal, homozygous deletions of the HIF1α locus on 14q in clear cell renal carcinoma cell lines. Wild-type HIF1α suppresses renal carcinoma growth, but the products of these altered loci do not. Conversely, downregulation of HIF1α in HIF1α-proficient lines promotes tumor growth. HIF1α activity is diminished in 14q-deleted kidney cancers, and all somatic HIF1α mutations identified in kidney cancers tested to date are loss of function. Therefore, HIF1α has the credentials of a kidney cancer suppressor gene. SIGNIFICANCE Deletion of 14q is a frequent event in clear cell renal carcinoma and portends a poor prognosis. In this study, we provide genetic and functional evidence that HIF1α is a target of 14q loss in kidney cancer.
Collapse
Affiliation(s)
- Chuan Shen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Broad Institute, Cambridge, MA 02142
| | - Steven E. Schumacher
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115
- Broad Institute, Cambridge, MA 02142
| | - Jing Zhou
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Michelle Chang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Broad Institute, Cambridge, MA 02142
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| |
Collapse
|
37
|
Nagao K, Oka K. HIF-2 directly activates CD82 gene expression in endothelial cells. Biochem Biophys Res Commun 2011; 407:260-5. [PMID: 21382346 DOI: 10.1016/j.bbrc.2011.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 03/03/2011] [Indexed: 10/18/2022]
Abstract
Hypoxia inducible factor (HIF)-1 and HIF-2 are transcription factors that mediate the cellular response to hypoxia. Although HIF-1 and HIF-2 share the same target genes, both proteins activate a distinct subset of genes. To identify the target genes preferentially activated by HIF-2 in endothelial cells, DNA microarray analysis was performed to human umbilical vein endothelial cells (HUVECs) with forced expression of either HIF-1α or HIF-2α. In the present study, which is the first comparative study of target genes induced by either HIF-1 or HIF-2 in HUVECs, HIF-1 (and not HIF-2) stimulated mainly glycolytic, hexose metabolic and alcohol metabolic gene expression. However, HIF-2 (but not HIF-1) induced developmental gene expressions such as Fms-like tyrosine kinase 1 (Flt-1) and angiopoietin 2 (Angpt2). Furthermore, CD82 was up-regulated by HIF-2, but not by HIF-1, in response to hypoxia. HIF-2 regulated CD82 gene expression by binding to its HRE consensus sequence located within its first intron. Assessing the function of CD82 in HUVECs forced its expression. This result revealed that CD82 negatively regulates the HUVECs cell migration. The induction of CD82 gene expression in endothelial cells provided new insights into a specific function of HIF-2.
Collapse
Affiliation(s)
- Kaori Nagao
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan.
| | | |
Collapse
|
38
|
Prabhu V, Guruvayoorappan C. Prolyl hydroxylase and hypoxia inducible factor: potential targets for cancer therapy. Immunopharmacol Immunotoxicol 2011; 33:568-75. [DOI: 10.3109/08923973.2010.545418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
39
|
Zhu GQ, Tang YL, Li L, Zheng M, Jiang J, Li XY, Chen SX, Liang XH. Hypoxia inducible factor 1α and hypoxia inducible factor 2α play distinct and functionally overlapping roles in oral squamous cell carcinoma. Clin Cancer Res 2010; 16:4732-41. [PMID: 20807755 DOI: 10.1158/1078-0432.ccr-10-1408] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE This study aimed to investigate the functional difference between hypoxia inducible factor (HIF)-1α and HIF-2α in oral squamous cell carcinomas (OSCC). EXPERIMENTAL DESIGN We evaluated the correlations between HIF-1α and HIF-2α expression and the clinical-pathologic characteristics of 97 patients with OSCC by immunohistochemical staining. OSCC cell lines transfected with lentivirus encoding short hairpin RNA against HIF-1α/2α were used to investigate the HIF-1α/2α-dependent target genes. Xenograft tumors in nude mice were established using cells affected by lentivirus, and tumor growth, angiogenesis, proliferation, and apoptosis were measured. RESULTS HIF-1α expression was significantly associated with T stage (P = 0.004), lymph node involvement (P = 0.006), histologic differentiation (P = 0.013), and microvessel density (P = 0.014), whereas that of HIF-2α was associated with T stage (P = 0.011) and microvessel density (P = 0.005). Patients with positive HIF-1α nuclear staining had a significantly worse overall survival (P < 0.001) and disease-free survival (P < 0.001) than those with negative HIF-1α staining. When OSCC cells were cultured at 5% O(2), only HIF-2α contributed to the expression of vascular endothelial growth factor. At 1% O(2), vascular endothelial growth factor was regulated by both HIF-1α and HIF-2α, but glucose transporter 1, carbonic anhydrase 9, and urokinase-type plasminogen activator receptor were regulated by HIF-1α rather than by HIF-2α. Knocking down HIF-1α or HIF-2α individually inhibited the xenograft tumor angiogenesis and growth, and knocking them down simultaneously revealed a better inhibitory effect than knocking down either unit alone. CONCLUSIONS HIF-1α and HIF-2α correlated with different clinical-pathologic parameters, stabilized at different oxygen levels, and regulated different genes in OSCC. However, both HIF-1α and HIF-2α showed promoting roles in tumor angiogenesis and growth, and therapeutic outcome may benefit from combined targeting of HIF-1α and HIF-2α.
Collapse
Affiliation(s)
- Gui-quan Zhu
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Knowles HJ, Schaefer KL, Dirksen U, Athanasou NA. Hypoxia and hypoglycaemia in Ewing's sarcoma and osteosarcoma: regulation and phenotypic effects of Hypoxia-Inducible Factor. BMC Cancer 2010; 10:372. [PMID: 20637078 PMCID: PMC2918574 DOI: 10.1186/1471-2407-10-372] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 07/16/2010] [Indexed: 11/15/2022] Open
Abstract
Background Hypoxia regulates gene expression via the transcription factor HIF (Hypoxia-Inducible Factor). Little is known regarding HIF expression and function in primary bone sarcomas. We describe HIF expression and phenotypic effects of hypoxia, hypoglycaemia and HIF in Ewing's sarcoma and osteosarcoma. Methods HIF-1α and HIF-2α immunohistochemistry was performed on a Ewing's tumour tissue array. Ewing's sarcoma and osteosarcoma cell lines were assessed for HIF pathway induction by Western blot, luciferase assay and ELISA. Effects of hypoxia, hypoglycaemia and isoform-specific HIF siRNA were assessed on proliferation, apoptosis and migration. Results 17/56 Ewing's tumours were HIF-1α-positive, 15 HIF-2α-positive and 10 positive for HIF-1α and HIF-2α. Expression of HIF-1α and cleaved caspase 3 localised to necrotic areas. Hypoxia induced HIF-1α and HIF-2α in Ewing's and osteosarcoma cell lines while hypoglycaemia specifically induced HIF-2α in Ewing's. Downstream transcription was HIF-1α-dependent in Ewing's sarcoma, but regulated by both isoforms in osteosarcoma. In both cell types hypoglycaemia reduced cellular proliferation by ≥ 45%, hypoxia increased apoptosis and HIF siRNA modulated hypoxic proliferation and migration. Conclusions Co-localisation of HIF-1α and necrosis in Ewing's sarcoma suggests a role for hypoxia and/or hypoglycaemia in in vivo induction of HIF. In vitro data implicates hypoxia as the primary HIF stimulus in both Ewing's and osteosarcoma, driving effects on proliferation and apoptosis. These results provide a foundation from which to advance understanding of HIF function in the pathobiology of primary bone sarcomas.
Collapse
Affiliation(s)
- Helen J Knowles
- Botnar Research Centre, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7LD, UK.
| | | | | | | |
Collapse
|
41
|
Prognostic and predictive biomarkers in renal cell carcinoma. Target Oncol 2010; 5:85-94. [DOI: 10.1007/s11523-010-0143-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/17/2010] [Indexed: 11/27/2022]
|
42
|
Biswas S, Troy H, Leek R, Chung YL, Li JL, Raval RR, Turley H, Gatter K, Pezzella F, Griffiths JR, Stubbs M, Harris AL. Effects of HIF-1alpha and HIF2alpha on Growth and Metabolism of Clear-Cell Renal Cell Carcinoma 786-0 Xenografts. JOURNAL OF ONCOLOGY 2010; 2010:757908. [PMID: 20652061 PMCID: PMC2905950 DOI: 10.1155/2010/757908] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 03/09/2010] [Accepted: 04/22/2010] [Indexed: 11/18/2022]
Abstract
In cultured clear-cell renal carcinoma (CCRCC) 786-0 cells transfected with HIF1alpha (HIF-1+), HIF-2alpha (HIF-2+), or empty vector (EV), no significant differences were observed in the growth rates in vitro, but when grown in vivo as xenografts HIF-2alpha significantly increased, and HIF-1alpha significantly decreased growth rates, compared to EV tumors. Factors associated with proliferation were increased and factors associated with cell death were decreased in HIF-2+ tumors. Metabolite profiles showed higher glucose and lower lactate and alanine levels in the HIF-2+ tumors whilst immunostaining demonstrated higher pyruvate dehydrogenase and lower pyruvate dehydrogenase kinase 1, compared to control tumors. Taken together, these results suggest that overexpression of HIF-2alpha in CCRCC 786-0 tumors regulated growth both by maintaining a low level of glycolysis and by allowing more mitochondrial metabolism and tolerance to ROS induced DNA damage. The growth profiles observed may be mediated by adaptive changes to a more oxidative phenotype.
Collapse
Affiliation(s)
- Swethajit Biswas
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- CR UK Tumour Pathology Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Northern Institute for Cancer Research (NICR), Newcastle University, Freeman Hospital, Newcastle-Upon-Tyne NE7 7DN, UK
| | - Helen Troy
- CR UK Biomedical Magnetic Resonance Research Group, Division of Basic Medical Sciences, St. George's, University of London, London SW17 0RE, UK
- CRUK Clinical Magnetic Resonance Research Group, Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Russell Leek
- CR UK Tumour Pathology Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Yuen-Li Chung
- CR UK Biomedical Magnetic Resonance Research Group, Division of Basic Medical Sciences, St. George's, University of London, London SW17 0RE, UK
- CRUK Clinical Magnetic Resonance Research Group, Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Ji-liang Li
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Raju R. Raval
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Helen Turley
- CR UK Tumour Pathology Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Kevin Gatter
- CR UK Tumour Pathology Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Francesco Pezzella
- CR UK Tumour Pathology Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - John R. Griffiths
- CR UK Biomedical Magnetic Resonance Research Group, Division of Basic Medical Sciences, St. George's, University of London, London SW17 0RE, UK
- CRUK Cambridge Research Centre, Li Ka Shing Centre, Robinson Way, Cambridge CB2 ORE, UK
| | - Marion Stubbs
- CR UK Biomedical Magnetic Resonance Research Group, Division of Basic Medical Sciences, St. George's, University of London, London SW17 0RE, UK
- CRUK Cambridge Research Centre, Li Ka Shing Centre, Robinson Way, Cambridge CB2 ORE, UK
| | - Adrian L. Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
43
|
Paulis YWJ, Soetekouw PMMB, Verheul HMW, Tjan-Heijnen VCG, Griffioen AW. Signalling pathways in vasculogenic mimicry. Biochim Biophys Acta Rev Cancer 2010; 1806:18-28. [PMID: 20079807 DOI: 10.1016/j.bbcan.2010.01.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/24/2009] [Accepted: 01/07/2010] [Indexed: 12/13/2022]
Abstract
Solid tumour growth is dependent on the development of an adequate blood supply. For years, sprouting angiogenesis has been considered an exclusive mechanism of tumour vascularization. However, over the last years, several other mechanisms have been identified, including vessel-co-option, intussusception, recruitment of endothelial precursor cells (EPCs) and even mechanisms that do not involve endothelial cells, a process called vasculogenic mimicry (VM). The latter describes a mechanism by which highly aggressive tumour cells can form vessel-like structures themselves, by virtue of their high plasticity. VM has been observed in several tumour types and its occurrence is strongly associated with a poor prognosis. This review will focus on signalling molecules and cascades involved in VM. In addition, we will discuss the presence of VM in relation to ongoing cancer research. Finally, we describe the clinical significance of VM regarding anti-angiogenesis treatment modalities.
Collapse
Affiliation(s)
- Yvette W J Paulis
- Department of Internal Medicine, Division of Medical Oncology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
44
|
Moreno-Manzano V, Rodríguez-Jiménez FJ, Aceña-Bonilla JL, Fustero-Lardíes S, Erceg S, Dopazo J, Montaner D, Stojkovic M, Sánchez-Puelles JM. FM19G11, a new hypoxia-inducible factor (HIF) modulator, affects stem cell differentiation status. J Biol Chem 2009; 285:1333-42. [PMID: 19897487 DOI: 10.1074/jbc.m109.008326] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The biology of the alpha subunits of hypoxia-inducible factors (HIFalpha) has expanded from their role in angiogenesis to their current position in the self-renewal and differentiation of stem cells. The results reported in this article show the discovery of FM19G11, a novel chemical entity that inhibits HIFalpha proteins that repress target genes of the two alpha subunits, in various tumor cell lines as well as in adult and embryonic stem cell models from rodents and humans, respectively. FM19G11 inhibits at nanomolar range the transcriptional and protein expression of Oct4, Sox2, Nanog, and Tgf-alpha undifferentiating factors, in adult rat and human embryonic stem cells, FM19G11 activity occurs in ependymal progenitor stem cells from rats (epSPC), a cell model reported for spinal cord regeneration, which allows the progression of oligodendrocyte cell differentiation in a hypoxic environment, has created interest in its characterization for pharmacological research. Experiments using small interfering RNA showed a significant depletion in Sox2 protein only in the case of HIF2alpha silencing, but not in HIF1alpha-mediated ablation. Moreover, chromatin immunoprecipitation data, together with the significant presence of functional hypoxia response element consensus sequences in the promoter region of Sox2, strongly validated that this factor behaves as a target gene of HIF2alpha in epSPCs. FM19G11 causes a reduction of overall histone acetylation with significant repression of p300, a histone acetyltransferase required as a co-factor for HIF-transcription activation. Arrays carried out in the presence and absence of the inhibitor showed the predominant involvement of epigenetic-associated events mediated by the drug.
Collapse
|
45
|
Webb JD, Coleman ML, Pugh CW. Hypoxia, hypoxia-inducible factors (HIF), HIF hydroxylases and oxygen sensing. Cell Mol Life Sci 2009; 66:3539-54. [PMID: 19756382 PMCID: PMC11115642 DOI: 10.1007/s00018-009-0147-7] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 08/20/2009] [Indexed: 01/08/2023]
Abstract
This article outlines the need for a homeostatic response to alterations in cellular oxygenation. It describes work on erythropoietin control that led to the discovery of the hypoxia-inducible transcription factor (HIF-1) and the parallel recognition that this system was responsive to a widespread oxygen-sensing mechanism. Subsequently, multiple HIF isoforms have been shown to have overlapping but non-redundant functions, controlling expression of genes involved in diverse processes such as angiogenesis, vascular tone, metal transport, glycolysis, mitochondrial function, cell growth and survival. The major role of prolyl and asparaginyl hydroxylation in regulating HIFs is described, as well as the identification of PHD1-3 and FIH as the oxygen-sensing enzymes responsible for these hydroxylations. Current understanding of other processes that modulate overall HIF activity, including influences from other signalling mechanisms such as kinases and nitric oxide levels, and the existence of a variety of feedback loops are outlined. The effects of some mutations in this pathway are documented as is knowledge of other substrates for these enzymes. The importance of PHD1-3 and FIH, and the large family of 2-oxoglutarate and iron(II)-dependent dioxygenases of which they are a part, in biology and medicine are discussed.
Collapse
Affiliation(s)
- James D. Webb
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7BN UK
| | - Mathew L. Coleman
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7BN UK
| | - Christopher W. Pugh
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7BN UK
| |
Collapse
|
46
|
Mazumdar J, Dondeti V, Simon MC. Hypoxia-inducible factors in stem cells and cancer. J Cell Mol Med 2009; 13:4319-28. [PMID: 19900215 PMCID: PMC2874971 DOI: 10.1111/j.1582-4934.2009.00963.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 10/28/2009] [Indexed: 12/21/2022] Open
Abstract
Cellular properties are influenced by complex factors inherent to their microenvironments. While oxygen deprivation (hypoxia) occurs in tumours because of rapid cell proliferation and aberrant blood vessel formation, embryonic cells develop in a naturally occurring hypoxic environment. Cells respond to hypoxia by stabilizing hypoxia-inducible factors (HIFs), which are traditionally viewed to function by altering cellular metabolism and blood vessel architecture. Recently, HIFs have been shown to modulate specific stem cell effectors, such as Notch, Wnt and Oct4 that control stem cell proliferation, differentiation and pluripotency. Direct molecular links have also been established between HIFs and critical cell signalling pathways such as cMyc and p53. These novel links suggest a new role for HIFs in stem cell and tumour regulation.
Collapse
Affiliation(s)
- Jolly Mazumdar
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine,Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - Vijay Dondeti
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine,Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine,Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| |
Collapse
|
47
|
Abstract
The central component of hypoxia sensing in the cell is the hypoxia-inducible factor (HIF) transcriptional complex. HIF activity is deregulated in many human cancers, especially those that are highly hypoxic. Hypoxic tumour cells are usually resistant to radiotherapy and most conventional chemotherapeutic agents, rendering them highly aggressive and metastatic. Overexpression of HIF-alpha, the regulatory subunit of HIF, is associated with increased vascular density, severity of tumour grade, treatment failure and a poor prognostic outcome with conventional therapies. Therefore HIF is an attractive, although challenging, therapeutic target, and several different strategies have been developed to target HIF directly or indirectly in recent years. This review outlines the preclinical and clinical advances in this arena and discusses which cancers may benefit from HIF-targeted therapy.
Collapse
|
48
|
Koh MY, Powis G. HAF : the new player in oxygen-independent HIF-1alpha degradation. Cell Cycle 2009; 8:1359-66. [PMID: 19377289 DOI: 10.4161/cc.8.9.8303] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Adaptation to hypoxia is primarily mediated by the hypoxia-inducible transcription factor, HIF. The regulation of HIF activity by the oxygen-dependent degradation of the HIF-1alpha and HIF-2alpha subunits by the pVHL E3 ligase complex has been well characterized. We have recently described the hypoxia-associated factor, HAF, as an E3 ligase for HIF-1alpha that does not degrade HIF-2alpha. Here we summarize the mechanism of HAF-mediated HIF-1alpha degradation and the importance of oxygen-independent HIF-1alpha regulation in cancer. We also discuss the implications of the new HAF: HIF-1alpha degradation pathway with respect to other novel mediators of oxygen-independent HIF-alpha degradation. Finally, we review the significance of HAF as an isoform-specific E3 ligase in light of new information on the non-overlapping functions of HIF-1alpha and HIF-2alpha in cancer.
Collapse
Affiliation(s)
- Mei Yee Koh
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
49
|
Bertout JA, Patel SA, Fryer BH, Durham AC, Covello KL, Olive KP, Goldschmidt MH, Simon MC. Heterozygosity for hypoxia inducible factor 1alpha decreases the incidence of thymic lymphomas in a p53 mutant mouse model. Cancer Res 2009; 69:3213-20. [PMID: 19293180 DOI: 10.1158/0008-5472.can-08-4223] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hypoxia inducible factors (HIF) are critical mediators of the cellular response to decreased oxygen tension and are overexpressed in a number of tumors. Although HIF1alpha and HIF2alpha share a high degree of sequence homology, recent work has shown that the two alpha subunits can have contrasting and tissue-specific effects on tumor growth. To directly compare the role of each HIFalpha subunit in spontaneous tumorigenesis, we bred a mouse model of expanded HIF2alpha expression and Hif1alpha(+/-) mice to homozygotes for the R270H mutation in p53. Here, we report that p53(R270H/R270H) mice, which have not been previously described, develop a unique tumor spectrum relative to p53(R270H/-) mice, including a high incidence of thymic lymphomas. Heterozygosity for Hif1alpha significantly reduced the incidence of thymic lymphomas observed in this model. Moreover, reduced Hif1alpha levels correlated with decreased stabilization of activated Notch1 and expression of the Notch target genes, Dtx1 and Nrarp. These observations uncover a novel role for HIF1alpha in Notch pathway activation during T-cell lymphomagenesis.
Collapse
Affiliation(s)
- Jessica A Bertout
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Von Hippel-Lindau (VHL) disease is caused by germline mutations in the VHL tumor suppressor gene, with Type 2B missense VHL mutations predisposing to renal cell carcinoma, hemangioblastoma and pheochromocytoma. Type 2B mutant pVHL is predicted to be defective in hypoxia inducible factor (HIF)-alpha regulation. Murine embryonic stem (ES) cells in which the endogenous wild-type Vhl gene was replaced with the representative Type 2B VHL hotspot mutation R167Q (Vhl(2B/2B)) displayed preserved physiological regulation of both HIF factors with slightly greater normoxic dysregulation of HIF-2alpha. Differentiated Vhl(2B/2B)-derived teratomas overexpressed joint HIF targets Vegf and EglN3 but not the HIF-1alpha-specific target Pfk1. Vhl(2B/2B) teratomas additionally displayed a growth advantage over Vhl(-/-)-derived teratomas, suggestive of a tight connection between perturbations in the degree and ratio of HIF-1alpha and HIF-2alpha stabilization and cell growth. Vhl(2B/2B) mice displayed mid-gestational embryonic lethality, whereas adult Vhl(2B/+) mice exhibited susceptibility to carcinogen-promoted renal neoplasia compared with wild-type littermates at 12 months. Our experiments support a model in which the representative Type 2B R167Q mutant pVhl produces a unique profile of HIF dysregulation, thereby promoting tissue-specific effects on cell growth, development and tumor predisposition.
Collapse
|