501
|
Julian CG. An Aptitude for Altitude: Are Epigenomic Processes Involved? Front Physiol 2019; 10:1397. [PMID: 31824328 PMCID: PMC6883803 DOI: 10.3389/fphys.2019.01397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/29/2019] [Indexed: 12/30/2022] Open
Abstract
In recent years, high-throughput genomic technologies and computational advancements have invigorated efforts to identify the molecular mechanisms regulating human adaptation to high altitude. Although exceptional progress regarding the identification of genomic regions showing evidence of recent positive selection has been made, many of the key “hypoxia tolerant” phenotypes of highland populations have not yet been linked to putative adaptive genetic variants. As a result, fundamental questions regarding the biological processes by which such adaptations are acquired remain unanswered. This Mini Review discusses the hypothesis that the epigenome works in coordination with underlying genomic sequence to govern adaptation to the chronic hypoxia of high altitude by influencing adaptive capacity and phenotypic variation under conditions of environmental hypoxia. Efforts to unravel the complex interactions between the genome, epigenome, and environmental exposures are essential to more fully appreciate the mechanisms underlying human adaptation to hypoxia.
Collapse
Affiliation(s)
- Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
502
|
Lenting K, van den Heuvel CNAM, van Ewijk A, ElMelik D, de Boer R, Tindall E, Wei G, Kusters B, te Dorsthorst M, ter Laan M, Huynen MA, Leenders WP. Mapping actionable pathways and mutations in brain tumours using targeted RNA next generation sequencing. Acta Neuropathol Commun 2019; 7:185. [PMID: 31747973 PMCID: PMC6865071 DOI: 10.1186/s40478-019-0826-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/02/2019] [Indexed: 01/28/2023] Open
Abstract
Many biology-based precision drugs are available that neutralize aberrant molecular pathways in cancer. Molecular heterogeneity and the lack of reliable companion diagnostic biomarkers for many drugs makes targeted treatment of cancer inaccurate for many individuals. Identifying actionable hyperactive biological pathways in individual cancers may improve this situation. To achieve this we applied a novel targeted RNA next generation sequencing (t/RNA-NGS) technique to surgically obtained glioma tissues. The test combines mutation detection with analysis of biological pathway activities that are involved in tumour behavior in many cancer types (e.g. tyrosine kinase signaling, angiogenesis signaling, immune response, metabolism), via quantitative measurement of transcript levels and splice variants of hundreds of genes. We here present proof of concept that the technique, which uses molecular inversion probes, generates a histology-independent molecular diagnosis and identifies classifiers that are strongly associated with conventional histopathology diagnoses and even with patient prognosis. The test not only confirmed known glioma-associated molecular aberrations but also identified aberrant expression levels of actionable genes and mutations that have so far been considered not to be associated with glioma, opening up the possibility of drug repurposing for individual patients. Its cost-effectiveness makes t/RNA-NGS to an attractive instrument to aid oncologists in therapy decision making.
Collapse
|
503
|
Zuniga-Hertz JP, Patel HH. The Evolution of Cholesterol-Rich Membrane in Oxygen Adaption: The Respiratory System as a Model. Front Physiol 2019; 10:1340. [PMID: 31736773 PMCID: PMC6828933 DOI: 10.3389/fphys.2019.01340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
The increase in atmospheric oxygen levels imposed significant environmental pressure on primitive organisms concerning intracellular oxygen concentration management. Evidence suggests the rise of cholesterol, a key molecule for cellular membrane organization, as a cellular strategy to restrain free oxygen diffusion under the new environmental conditions. During evolution and the increase in organismal complexity, cholesterol played a pivotal role in the establishment of novel and more complex functions associated with lipid membranes. Of these, caveolae, cholesterol-rich membrane domains, are signaling hubs that regulate important in situ functions. Evolution resulted in complex respiratory systems and molecular response mechanisms that ensure responses to critical events such as hypoxia facilitated oxygen diffusion and transport in complex organisms. Caveolae have been structurally and functionally associated with respiratory systems and oxygen diffusion control through their relationship with molecular response systems like hypoxia-inducible factors (HIF), and particularly as a membrane-localized oxygen sensor, controlling oxygen diffusion balanced with cellular physiological requirements. This review will focus on membrane adaptations that contribute to regulating oxygen in living systems.
Collapse
Affiliation(s)
- Juan Pablo Zuniga-Hertz
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
504
|
Wang X, Li L, Zhao K, Lin Q, Li H, Xue X, Ge W, He H, Liu D, Xie H, Wu Q, Hu Y. A novel LncRNA HITT forms a regulatory loop with HIF-1α to modulate angiogenesis and tumor growth. Cell Death Differ 2019; 27:1431-1446. [PMID: 31700144 DOI: 10.1038/s41418-019-0449-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 11/09/2022] Open
Abstract
Increasing evidence has indicated that long noncoding RNAs (lncRNAs) play important roles in human diseases, including cancer; however, only a few of them have been experimentally validated and functionally annotated. Here, we identify a novel lncRNA that we term HITT (HIF-1α inhibitor at translation level). HITT is commonly decreased in multiple human cancers. Decreased HITT is associated with advanced stages of colon cancer. Restoration of the expression of HITT in cancer cells inhibits angiogenesis and tumor growth in vivo in an HIF-1α-dependent manner. Further study reveals that HITT inhibits HIF-1α expression, mainly by interfering with its translation. Mechanically, HITT titrates away YB-1 from the 5'-UTR of HIF-1α mRNA via a high-stringency YB-1-binding motif. The reverse correlation between HITT and HIF-1α expression is further validated in human colon cancer tissues. Moreover, HITT is one of the most altered lncRNAs upon the hypoxic switch and HITT downregulation is required for hypoxia-induced HIF-1α expression. We further demonstrate that HITT and HIF-1α form an autoregulatory feedback loop where HIF-1α destabilizes HITT by inducing MiR-205, which directly targets HITT for degradation. Together, these results expand our understanding of the cancer-associated functions of lncRNAs, highlighting the HITT-HIF-1α axis as constituting an additional layer of regulation of angiogenesis and tumor growth, with potential implications for therapeutic targeting.
Collapse
Affiliation(s)
- Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Li Li
- The Affiliated Tumor Hospital of Harbin Medical University, 150001, Harbin, Heilongjiang Province, China
| | - Kunming Zhao
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Huayi Li
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Xuting Xue
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Wenjie Ge
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Hongjuan He
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Dong Liu
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Hui Xie
- State Key Laboratory of Robotics and Systems, Harbin Institute of Technology, 2 Yikuang, 150001, Harbin, China
| | - Qiong Wu
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, 150001, Harbin, Heilongjiang Province, China. .,Shenzhen Graduate School of Harbin Institute of Technology, 518055, Shenzhen, China.
| |
Collapse
|
505
|
Liu Y, Huang X, Yu H, Yang J, Li Y, Yuan X, Guo Q. HIF-1α-TWIST pathway restrains cyclic mechanical stretch-induced osteogenic differentiation of bone marrow mesenchymal stem cells. Connect Tissue Res 2019; 60:544-554. [PMID: 30938209 DOI: 10.1080/03008207.2019.1601185] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Aim: Mechanical strain plays a crucial role in bone formation and remodeling. Hypoxia-inducible factor (HIF)-1α and TWIST are upstream of master regulators of osteogenesis, including runt-related transcription factor 2 (RUNX2) and bone morphogenetic proteins (BMPs). This study investigated the effect of the HIF-1α-TWIST pathway on cyclic mechanical stretch-induced osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) and the underlying mechanism. Materials and Methods: BMSCs were isolated from bone marrow derived from the femurs and humeri of Sprague-Dawley rats. Osteogenic differentiation of BMSCs was induced by applying cyclic mechanical stretch using the Flexcell Tension System. HIF-1α and TWIST were knocked down using recombinant lentiviral vectors. Osteogenic differentiation was evaluated by real-time qPCR, western blotting, and the alkaline phosphatase (ALP) activity assay. Results: Cyclic mechanical stretch increased ALP activity and expression of HIF-1α and TWIST in BMSCs. Knockdown of HIF-1α decreased TWIST expression in stretched BMSCs. Moreover, knockdown of HIF-1α or TWIST enhanced cyclic mechanical stretch-induced osteogenic differentiation of BMSCs. In addition, knockdown of TWIST increased expression of RUNX2 and BMP2 in stretched BMSCs. Conclusions: The HIF-1α-TWIST signaling pathway inhibits cyclic mechanical stretch-induced osteogenic differentiation of BMSCs. This finding may facilitate cell and tissue engineering for clinical applications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China.,Department of Orthodontics, Stomatology College of Qingdao University , Qingdao , Shandong , China.,Department of Stomatology, The Affiliated Qingdao Municipal Hospital, Qingdao University , Qingdao , Shandong , China
| | - Xia Huang
- Department of Nursing and Hospital Infection Management, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Haibo Yu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Jing Yang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Yazhen Li
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Xiao Yuan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Qingyuan Guo
- Department of Stomatology, The Affiliated Qingdao Municipal Hospital, Qingdao University , Qingdao , Shandong , China.,Department of Stomatology, Chinese PLA General Hospital , Beijing , China
| |
Collapse
|
506
|
Fasano C, Disciglio V, Bertora S, Lepore Signorile M, Simone C. FOXO3a from the Nucleus to the Mitochondria: A Round Trip in Cellular Stress Response. Cells 2019; 8:cells8091110. [PMID: 31546924 PMCID: PMC6769815 DOI: 10.3390/cells8091110] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/25/2022] Open
Abstract
Cellular stress response is a universal mechanism that ensures the survival or negative selection of cells in challenging conditions. The transcription factor Forkhead box protein O3 (FOXO3a) is a core regulator of cellular homeostasis, stress response, and longevity since it can modulate a variety of stress responses upon nutrient shortage, oxidative stress, hypoxia, heat shock, and DNA damage. FOXO3a activity is regulated by post-translational modifications that drive its shuttling between different cellular compartments, thereby determining its inactivation (cytoplasm) or activation (nucleus and mitochondria). Depending on the stress stimulus and subcellular context, activated FOXO3a can induce specific sets of nuclear genes, including cell cycle inhibitors, pro-apoptotic genes, reactive oxygen species (ROS) scavengers, autophagy effectors, gluconeogenic enzymes, and others. On the other hand, upon glucose restriction, 5′-AMP-activated protein kinase (AMPK) and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) -dependent FOXO3a mitochondrial translocation allows the transcription of oxidative phosphorylation (OXPHOS) genes, restoring cellular ATP levels, while in cancer cells, mitochondrial FOXO3a mediates survival upon genotoxic stress induced by chemotherapy. Interestingly, these target genes and their related pathways are diverse and sometimes antagonistic, suggesting that FOXO3a is an adaptable player in the dynamic homeostasis of normal and stressed cells. In this review, we describe the multiple roles of FOXO3a in cellular stress response, with a focus on both its nuclear and mitochondrial functions.
Collapse
Affiliation(s)
- Candida Fasano
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Vittoria Disciglio
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Stefania Bertora
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Martina Lepore Signorile
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy.
| | - Cristiano Simone
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari Aldo Moro, 70124 Bari, Italy.
| |
Collapse
|
507
|
Piñeiro Fernández J, Luddy KA, Harmon C, O'Farrelly C. Hepatic Tumor Microenvironments and Effects on NK Cell Phenotype and Function. Int J Mol Sci 2019; 20:E4131. [PMID: 31450598 PMCID: PMC6747260 DOI: 10.3390/ijms20174131] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
The liver is a complex organ with critical physiological functions including metabolism, glucose storage, and drug detoxification. Its unique immune profile with large numbers of cytotoxic CD8+ T cells and significant innate lymphoid population, including natural killer cells, γ δ T cells, MAIT cells, and iNKTcells, suggests an important anti-tumor surveillance role. Despite significant immune surveillance in the liver, in particular large NK cell populations, hepatic cell carcinoma (HCC) is a relatively common outcome of chronic liver infection or inflammation. The liver is also the second most common site of metastatic disease. This discordance suggests immune suppression by the environments of primary and secondary liver cancers. Classic tumor microenvironments (TME) are poorly perfused, leading to accumulation of tumor cell metabolites, diminished O2, and decreased nutrient levels, all of which impact immune cell phenotype and function. Here, we focus on changes in the liver microenvironment associated with tumor presence and how they affect NK function and phenotype.
Collapse
Affiliation(s)
| | - Kimberly A Luddy
- School of Biochemistry and Immunology, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33626, USA.
| | - Cathal Harmon
- Brigham and Women's Hospital, Harvard Institutes of Medicine, Harvard Medical School, Boston, MA 02138, USA
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland.
| |
Collapse
|
508
|
A Cell Density-Dependent Reporter in the Drosophila S2 Cells. Sci Rep 2019; 9:11868. [PMID: 31413273 PMCID: PMC6694118 DOI: 10.1038/s41598-019-47652-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/22/2019] [Indexed: 01/20/2023] Open
Abstract
Cell density regulates many aspects of cell properties and behaviors including metabolism, growth, cytoskeletal structure and locomotion. Importantly, the responses by cultured cells to density signals also uncover key mechanisms that govern animal development and diseases in vivo. Here we characterized a density-responsive reporter system in transgenic Drosophila S2 cells. We show that the reporter genes are strongly induced in a cell density-dependent and reporter-independent fashion. The rapid and reversible induction occurs at the level of mRNA accumulation. We show that multiple DNA elements within the transgene sequences, including a metal response element from the metallothionein gene, contribute to the reporter induction. The reporter induction correlates with changes in multiple cell density and growth regulatory pathways including hypoxia, apoptosis, cell cycle and cytoskeletal organization. Potential applications of such a density-responsive reporter will be discussed.
Collapse
|
509
|
Hafidi ME, Buelna-Chontal M, Sánchez-Muñoz F, Carbó R. Adipogenesis: A Necessary but Harmful Strategy. Int J Mol Sci 2019; 20:ijms20153657. [PMID: 31357412 PMCID: PMC6696444 DOI: 10.3390/ijms20153657] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is considered to significantly increase the risk of the development of a vast range of metabolic diseases. However, adipogenesis is a complex physiological process, necessary to sequester lipids effectively to avoid lipotoxicity in other tissues, like the liver, heart, muscle, essential for maintaining metabolic homeostasis and has a crucial role as a component of the innate immune system, far beyond than only being an inert mass of energy storage. In pathophysiological conditions, adipogenesis promotes a pro-inflammatory state, angiogenesis and the release of adipokines, which become dangerous to health. It results in a hypoxic state, causing oxidative stress and the synthesis and release of harmful free fatty acids. In this review, we try to explain the mechanisms occurring at the breaking point, at which adipogenesis leads to an uncontrolled lipotoxicity. This review highlights the types of adipose tissue and their functions, their way of storing lipids until a critical point, which is associated with hypoxia, inflammation, insulin resistance as well as lipodystrophy and adipogenesis modulation by Krüppel-like factors and miRNAs.
Collapse
Affiliation(s)
- Mohammed El Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Mabel Buelna-Chontal
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico.
| |
Collapse
|
510
|
He H, Qie S, Guo Q, Chen S, Zou C, Lu T, Su Y, Zong J, Xu H, He D, Xu Y, Chen B, Pan J, Sang N, Lin S. Stanniocalcin 2 (STC2) expression promotes post-radiation survival, migration and invasion of nasopharyngeal carcinoma cells. Cancer Manag Res 2019; 11:6411-6424. [PMID: 31372045 PMCID: PMC6636319 DOI: 10.2147/cmar.s197607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/06/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Stanniocalcin 2 (STC2) expression is upregulated under multiple stress conditions including hypoxia, nutrient starvation and radiation. Overexpression of STC2 correlates with tumor progression and poor prognosis. Purpose: We previously demonstrated that overexpression of STC2 in nasopharyngeal carcinomas (NPC) positively correlates with radiation resistance and tumor metastasis, two major clinical obstacles to the improvement of NPC management. However, it remains elusive whether STC2 expression is a critical contributing factor for post-radiation survival and metastasis of NPC cells. Materials and methods: Using the radiation resistant CNE2 cell line as a model, we examined the importance of STC2 expression for post-radiation survival, migration and invasion. Here, we report the establishment of STC2 knockout lines (CNE2-STC2-KO) using the CRISPR/Cas9-based genome editing technique. Results: Compared with the parental line, STC2-KO cells showed similar proliferation and morphology in normal culture conditions, and loss of STC2 did not compromise the cell tumorigenicity in nude mice model. However, STC2-KO lines demonstrated increased sensitivity to X-radiation under either normoxic or hypoxic conditions. Particularly, upon X-radiation, parental CNE2 cells only slightly whereas STC2-KO cells remarkably decreased the migration and invasion ability. Cell cycle analysis revealed that loss of STC2 accumulated cells in G1 and G2/M phases but decreased S-population. Conclusion: These data indicate that the expression of STC2, which can be stimulated by metabolic or therapeutic stresses, is one important factor to promote survival and metastasis of post-radiation NPC cells. Therefore, targeting STC2 or relative downstream pathways may provide novel strategies to overcome radiation resistance and metastasis of NPC.
Collapse
Affiliation(s)
- Huocong He
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuo Qie
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Qiaojuan Guo
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuyang Chen
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Changyan Zou
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Tianzhu Lu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Ying Su
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jingfeng Zong
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Hanchuan Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Dan He
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Yun Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Bijuan Chen
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Nianli Sang
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Shaojun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
511
|
Epigenetics: A Potential Mechanism Involved in the Pathogenesis of Various Adverse Consequences of Obstructive Sleep Apnea. Int J Mol Sci 2019; 20:ijms20122937. [PMID: 31208080 PMCID: PMC6627863 DOI: 10.3390/ijms20122937] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/09/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Epigenetics is defined as the heritable phenotypic changes which do not involve alterations in the DNA sequence, including histone modifications, non-coding RNAs, and DNA methylation. Recently, much attention has been paid to the role of hypoxia-mediated epigenetic regulation in cancer, pulmonary hypertension, adaptation to high altitude, and cardiorenal disease. In contrast to sustained hypoxia, chronic intermittent hypoxia with re-oxygenation (IHR) plays a major role in the pathogenesis of various adverse consequences of obstructive sleep apnea (OSA), resembling ischemia re-perfusion injury. Nevertheless, the role of epigenetics in the pathogenesis of OSA is currently underexplored. This review proposes that epigenetic processes are involved in the development of various adverse consequences of OSA by influencing adaptive potential and phenotypic variability under conditions of chronic IHR. Improved understanding of the interaction between genetic and environmental factors through epigenetic regulations holds great value to give deeper insight into the mechanisms underlying IHR-related low-grade inflammation, oxidative stress, and sympathetic hyperactivity, and clarify their implications for biomedical research.
Collapse
|
512
|
Involvement of E3 Ligases and Deubiquitinases in the Control of HIF-α Subunit Abundance. Cells 2019; 8:cells8060598. [PMID: 31208103 PMCID: PMC6627837 DOI: 10.3390/cells8060598] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
The ubiquitin and hypoxia-inducible factor (HIF) pathways are cellular processes involved in the regulation of a variety of cellular functions. Enzymes called ubiquitin E3 ligases perform protein ubiquitylation. The action of these enzymes can be counteracted by another group of enzymes called deubiquitinases (DUBs), which remove ubiquitin from target proteins. The balanced action of these enzymes allows cells to adapt their protein content to a variety of cellular and environmental stress factors, including hypoxia. While hypoxia appears to be a powerful regulator of the ubiquitylation process, much less is known about the impact of DUBs on the HIF system and hypoxia-regulated DUBs. Moreover, hypoxia and DUBs play crucial roles in many diseases, such as cancer. Hence, DUBs are considered to be promising targets for cancer cell-specific treatment. Here, we review the current knowledge about the role DUBs play in the control of HIFs, the regulation of DUBs by hypoxia, and their implication in cancer progression.
Collapse
|
513
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a brief summary about the current state of knowledge regarding the circadian rhythm in the regulation of normal renal function. RECENT FINDINGS There is a lack of information regarding how the circadian clock mechanisms may contribute to the development of diabetic kidney disease. We discuss recent findings regarding mechanisms that are established in diabetic kidney disease and are known to be linked to the circadian clock as possible connections between these two areas. Here, we hypothesize various mechanisms that may provide a link between the clock mechanism and kidney disease in diabetes based on available data from humans and rodent models.
Collapse
Affiliation(s)
- Olanrewaju A Olaoye
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
| | - Sarah H Masten
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
| | - Rajesh Mohandas
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, 1600 SW Archer Road, Box 100224, Gainesville, FL, 32610, USA.
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
514
|
Hypoxia induces the dormant state in oocytes through expression of Foxo3. Proc Natl Acad Sci U S A 2019; 116:12321-12326. [PMID: 31147464 DOI: 10.1073/pnas.1817223116] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In mammals, most immature oocytes remain dormant in the primordial follicles to ensure the longevity of female reproductive life. A precise understanding of mechanisms underlying the dormancy is important for reproductive biology and medicine. In this study, by comparing mouse oogenesis in vivo and in vitro, the latter of which bypasses the primordial follicle stage, we defined the gene-expression profile representing the dormant state of oocytes. Overexpression of constitutively active FOXO3 partially reproduced the dormant state in vitro. Based on further gene-expression analysis, we found that a hypoxic condition efficiently induced the dormant state in vitro. The effect of hypoxia was severely diminished by disruption of the Foxo3 gene and inhibition of hypoxia-inducible factors. Our findings provide insights into the importance of environmental conditions and their effectors for establishing the dormant state.
Collapse
|
515
|
Oxygen Regulates Human Pluripotent Stem Cell Metabolic Flux. Stem Cells Int 2019; 2019:8195614. [PMID: 31236115 PMCID: PMC6545818 DOI: 10.1155/2019/8195614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolism has been shown to alter cell fate in human pluripotent stem cells (hPSC). However, current understanding is almost exclusively based on work performed at 20% oxygen (air), with very few studies reporting on hPSC at physiological oxygen (5%). In this study, we integrated metabolic, transcriptomic, and epigenetic data to elucidate the impact of oxygen on hPSC. Using 13C-glucose labeling, we show that 5% oxygen increased the intracellular levels of glycolytic intermediates, glycogen, and the antioxidant response in hPSC. In contrast, 20% oxygen increased metabolite flux through the TCA cycle, activity of mitochondria, and ATP production. Acetylation of H3K9 and H3K27 was elevated at 5% oxygen while H3K27 trimethylation was decreased, conforming to a more open chromatin structure. RNA-seq analysis of 5% oxygen hPSC also indicated increases in glycolysis, lysine demethylases, and glucose-derived carbon metabolism, while increased methyltransferase and cell cycle activity was indicated at 20% oxygen. Our findings show that oxygen drives metabolite flux and specifies carbon fate in hPSC and, although the mechanism remains to be elucidated, oxygen was shown to alter methyltransferase and demethylase activity and the global epigenetic landscape.
Collapse
|
516
|
Yang W, Liu Y, Gao R, Xiu Z, Sun T. Knockdown of cZNF292 suppressed hypoxic human hepatoma SMMC7721 cell proliferation, vasculogenic mimicry, and radioresistance. Cell Signal 2019; 60:122-135. [PMID: 31028816 DOI: 10.1016/j.cellsig.2019.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 02/08/2023]
Abstract
Hypoxia is a classic feature of the tumor microenvironment, and has been established as a key epigenetic factor modulating the outcome of radiotherapy. Circular RNAs (circRNAs) are novel RNA molecules with covalently closed circular structures and are highly expressed in eukaryotic transcriptomes. Although previous analysis have shown that circRNA ZNF292 (cZNF292) was hypoxia-responsive and exhibited a proangiogenic function in vitro, the molecular mechanism of cZNF292's biological function is still unclear and deserves further exploration. In this study, we investigated the effect of cZNF292 on the vasculogenic mimicry (VM) and radiosensitivity of hypoxic hepatoma SMMC7721 cells and its mechanism. Our data indicated that cZNF292 could be induced by hypoxia in a time-dependent manner in hepatoma cells independent of hypoxia inducible factor (HIF)-1α. Knockdown of cZNF292 increased SRY (sex determining region Y)-box 9 (SOX9) nuclear translocation, subsequently reduced Wnt/β-catenin pathway activity, leading to suppression of hypoxic hepatoma cell proliferation, VM, and radioresistance in vitro and in vivo. Our results delineated a novel mechanism of cZNF292 in enhancing hypoxic tumor cell radiosensitivity, which might provide valuable targets for radiation therapy for hepatoma.
Collapse
Affiliation(s)
- Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China.
| | - Yingying Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China; Isotopic Laboratory of Nuclear Medicine, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Ruoling Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Zenghe Xiu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Ting Sun
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
517
|
Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Han HJ. Role of HIF1 α Regulatory Factors in Stem Cells. Int J Stem Cells 2019; 12:8-20. [PMID: 30836734 PMCID: PMC6457711 DOI: 10.15283/ijsc18109] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF1) is a master transcription factor that induces the transcription of genes involved in the metabolism and behavior of stem cells. HIF1-mediated adaptation to hypoxia is required to maintain the pluripotency and survival of stem cells under hypoxic conditions. HIF1 activity is well known to be tightly controlled by the alpha subunit of HIF1 (HIF1α). Understanding the regulatory mechanisms that control HIF1 activity in stem cells will provide novel insights into stem cell biology under hypoxia. Recent research has unraveled the mechanistic details of HIF1α regulating processes, suggesting new strategies for regulating stem cells. This review summarizes recent experimental studies on the role of several regulatory factors (including calcium, 2-oxoglutarate-dependent dioxygenase, microtubule network, importin, and coactivators) in regulating HIF1α activity in stem cells.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| |
Collapse
|
518
|
Hayashi Y, Yokota A, Harada H, Huang G. Hypoxia/pseudohypoxia-mediated activation of hypoxia-inducible factor-1α in cancer. Cancer Sci 2019; 110:1510-1517. [PMID: 30844107 PMCID: PMC6501028 DOI: 10.1111/cas.13990] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Since the first identification of hypoxic cells in sections of carcinomas in the 1950s, hypoxia has been known as a central hallmark of cancer cells and their microenvironment. Indeed, hypoxia benefits cancer cells in their growth, survival, and metastasis. The historical discovery of hypoxia‐inducible factor‐1α (HIF1A) in the early 1990s had a great influence on the field as many phenomena in hypoxia could be explained by HIF1A. However, not all regions or types of tumors are necessarily hypoxic. Thus, it is difficult to explain whole cancer pathobiology by hypoxia, especially in the early stage of cancer. Upregulation of glucose metabolism in cancer cells has been well known. Oxygen‐independent glycolysis is activated in cancer cells even in the normoxia condition, which is known as the Warburg effect. Accumulating evidence and recent advances in cancer metabolism research suggest that hypoxia‐independent mechanisms for HIF signaling activation is a hallmark for cancer. There are various mechanisms that generate pseudohypoxic conditions, even in normoxia. Given the importance of HIF1A for cancer pathobiology, the pseudohypoxia concept could shed light on the longstanding mystery of the Warburg effect and accelerate better understanding of the diverse phenomena seen in a variety of cancers.
Collapse
Affiliation(s)
- Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Asumi Yokota
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
519
|
Qian J, Gelens L, Bollen M. Coordination of Timers and Sensors in Cell Signaling. Bioessays 2019; 41:e1800217. [PMID: 30730051 DOI: 10.1002/bies.201800217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/12/2018] [Indexed: 02/06/2023]
Abstract
Timers and sensors are common devices that make our daily life safer, more convenient, and more efficient. In a cellular context, they arguably play an even more crucial role as they ensure the survival of cells in the presence of various extrinsic and intrinsic stresses. Biological timers and sensors generate distinct signaling profiles, enabling them to produce different types of cellular responses. Recent data suggest that they can work together to guarantee correct timing and responsiveness. By exploring examples of cellular stress signaling from mitosis, DNA damage, and hypoxia, the authors discuss the common architecture of timer-sensor integration, and how its added features contribute to the generation of desired signaling profiles when dealing with stresses of variable duration and strength. The authors propose timer-sensor integration as a widespread mechanism with profound biological implications and therapeutic potential.
Collapse
Affiliation(s)
- Junbin Qian
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium.,VIB Center for Cancer Biology, 3000, Leuven, Belgium.,Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Lendert Gelens
- Laboratory of Dynamics in Biological Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Mathieu Bollen
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
520
|
Hou Y, Wang X, Chen X, Zhang J, Ai X, Liang Y, Yu Y, Zhang Y, Meng X, Kuang T, Hu Y. Establishment and evaluation of a simulated high‑altitude hypoxic brain injury model in SD rats. Mol Med Rep 2019; 19:2758-2766. [PMID: 30720143 PMCID: PMC6423628 DOI: 10.3892/mmr.2019.9939] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022] Open
Abstract
This study was conducted to establish a stable hypobaric hypoxia brain injury model. SD rats were randomly separated into control and model groups, and placed outside or inside of a hypobaric chamber, respectively. Subsequent to 24 h anoxic exposure, plasma superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG) and lactate dehydrogenase (LDH) were measured using commercial biochemical kits. Hematoxylin-eosin (H&E), Nissl's and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were used to observe the morphology of neurons in the hippocampus. The protein expression levels of apoptotic protease activating factor-1 (Apaf-1), hypoxia inducible factor-1α (HIF-1α), caspase-3, cleaved caspase-3, Bcl-2-associated X protein (Bax) and cytochrome c (cyto-c) were detected using western blot and immunohistochemistry analyses. Hypoxic substantially induced morphological lesions in the hippocampus concomitant with the physical behavioral performance deficit. Furthermore, hypoxia markedly exacerbated the levels of MDA, LDH and GSSG, and restrained GSH (P<0.01) and SOD (P<0.05) levels compared with the control group. In addition, hypoxia significantly induced the protein expression of Apaf-1, HIF-1α, caspase-3, cleaved caspase-3, Bax and Cyto-c (P<0.01) compared with the control group. Finally, a lower number and volume of Nissl bodies were verified in the hypoxic group. TUNEL results demonstrated a greater number of apoptotic cells in the hypoxic group. The present study demonstrates a model of rat hypoxic brain injuries induced by a hypobaric chamber at 9,000 m for 24 h. Furthermore, the redox enzyme, HIF-1α and mitochondrial apoptosis-associated protein, along with H&E and Nissl's staining, may be applied to evaluate the degree of injury.
Collapse
Affiliation(s)
- Ya Hou
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaobo Wang
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaorui Chen
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Jing Zhang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaopeng Ai
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yusheng Liang
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yangyang Yu
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yi Zhang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xianli Meng
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Tingting Kuang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yao Hu
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| |
Collapse
|
521
|
HIF-1α inducing exosomal microRNA-23a expression mediates the cross-talk between tubular epithelial cells and macrophages in tubulointerstitial inflammation. Kidney Int 2019; 95:388-404. [DOI: 10.1016/j.kint.2018.09.013] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/24/2018] [Accepted: 09/06/2018] [Indexed: 02/04/2023]
|
522
|
|
523
|
Polysaccharides from the Edible Mushroom Agaricus bitorquis (Quél.) Sacc. Chaidam Show Anti-hypoxia Activities in Pulmonary Artery Smooth Muscle Cells. Int J Mol Sci 2019; 20:ijms20030637. [PMID: 30717240 PMCID: PMC6387285 DOI: 10.3390/ijms20030637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 01/02/2023] Open
Abstract
Three kinds of new water-soluble polysaccharides (FA, FB and FC) were isolated from wild mushroom Agaricus bitorquis (Quél.) Sacc. Chaidam by the classical method “water extraction and alcohol precipitation” and purified by column chromatography. The Mw of FA, FB and FC ranged from 5690 Da to 38,340 Da. The three polysaccharide fractions in the fruiting body were mainly composed of 4 kinds of monosaccharides, including glucose, galactose, mannose, and arabinose, among which glucose and galactose were the major monosaccharides. The FTIR and NMR spectroscopy indicated that the skeleton of three fractions composed of a (1→4)-α-D-glycosidic backbone containing α-D-mannopyranose. In vitro anti-hypoxia activity data showed that three polysaccharide fractions possessed a significant effect on inhibiting PASM cells apoptosis under hypoxia. Among them, FC at the concentration of 200 µg/mL revealed a significant anti-hypoxia effect. These results revealed that the intracellular polysaccharides possessed potent anti-hypoxic activity, which might be related to inhibiting LDH and NADPH oxidase expression and promoting the formation of 5-hydroxytryptamine, dopamine, endothelins, acetylcholine. More importantly, FC showed good performance inducing KV1.5 expression and prohibiting KIR6.2 formation at protein level.
Collapse
|
524
|
Braicu C, Gulei D, Cojocneanu R, Raduly L, Jurj A, Knutsen E, Calin GA, Berindan‐Neagoe I. miR-181a/b therapy in lung cancer: reality or myth? Mol Oncol 2019; 13:9-25. [PMID: 30548184 PMCID: PMC6322195 DOI: 10.1002/1878-0261.12420] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Despite substantial progress in oncology, lung cancer remains the number one malignancy in terms of both incidence and mortality rates, and there thus remains an urgent need for new therapeutic alternatives. MicroRNA (miRNA) have an important role in cancer initiation and progression due to their capacity to interfere with transcriptional signaling and regulate key cellular processes. miR-181a and miR-181b (miR-181a/b), which are located on chromosomes 1 and 9, are pathologically expressed in the tumor tissue and plasma of patients diagnosed with lung cancer. The miR-181a/b regulatory mechanisms are sophisticated and are directly related to different target genes. In recent years, an ever-increasing number of studies have focused on the biological relevance of miR-181a/b in key cellular processes. In this paper, we aim to discuss the challenging experimental data related to miR-181a/b and their potential use for the development of new therapeutic approaches in lung cancer. We will further present the ongoing issues regarding the regulation of their multiple target genes, and their potential use as biomarkers and therapeutic targets in this deadly malignancy.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
| | - Diana Gulei
- MedFuture Research Center for Advanced Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
| | - Roxana Cojocneanu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
| | - Erik Knutsen
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - George Adrian Calin
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Center for RNA Inference and Non‐Coding RNAThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ioana Berindan‐Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
- MedFuture Research Center for Advanced Medicine‘Iuliu Hatieganu’ University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Functional Genomics and Experimental PathologyThe Oncology Institute ‘Prof. Dr. Ion Chiricuta’Cluj‐NapocaRomania
| |
Collapse
|
525
|
Su Y, Yang W, Jiang N, Shi J, Chen L, Zhong G, Bi J, Dong W, Wang Q, Wang C, Lin T. Hypoxia-elevated circELP3 contributes to bladder cancer progression and cisplatin resistance. Int J Biol Sci 2019; 15:441-452. [PMID: 30745833 PMCID: PMC6367558 DOI: 10.7150/ijbs.26826] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/18/2018] [Indexed: 12/22/2022] Open
Abstract
Hypoxia plays a critical role in cancer biology. It induces genomic instability, which in turn helps cancer cells respond adaptively to meet the needs of carcinogenesis, cancer progression and relapse. Circular RNA has not been reported among the variety of downstream factors in this adaptive response. Although a few studies have demonstrated the important role of circular RNAs in driving human bladder cancer progression, their carcinogenic roles are still under investigated. Here, we identified a hypoxia-elevated circular RNA, circELP3, that contributes to bladder cancer progression and cisplatin resistance. Decreasing the level of circELP3 via siRNA clearly reduced the in vitro proliferation and cisplatin resistance of bladder cancer cells and promoted apoptosis. Interfering with circELP3 suppressed tumor xenograft growth in nude mice in vivo. In addition, lower circELP3-expressing bladder cancer cells displayed poorer self-renewal capacity, as demonstrated by lower levels of sphere formation and stem cell marker expression. Furthermore, in human bladder cancer patients, strong correlations between a high circELP3 level and advanced tumor grade and lymph node metastasis were observed. In summary, we provide the first direct evidence that circular RNA participates in the adaptive response to hypoxia and may play a role in the progression and drug resistance of bladder cancer.
Collapse
Affiliation(s)
- Yinjie Su
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Weiping Yang
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,The Department of Urology, Yan'an Hospital Affiliated with Kunming Medical University, Kunming, China
| | - Ning Jiang
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,The Department of Urology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Juanyi Shi
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Luping Chen
- The Department of Pediatric Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guangzheng Zhong
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junming Bi
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei Dong
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiong Wang
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunhui Wang
- The Department of Urology, Yan'an Hospital Affiliated with Kunming Medical University, Kunming, China
| | - Tianxin Lin
- The Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
526
|
Wang D, Zhang X, Lu Y, Wang X, Zhu L. Hypoxia inducible factor 1α in hepatocellular carcinoma with cirrhosis: Association with prognosis. Pathol Res Pract 2018; 214:1987-1992. [DOI: 10.1016/j.prp.2018.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/23/2018] [Accepted: 09/11/2018] [Indexed: 01/16/2023]
|
527
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
528
|
BICD1 mediates HIF1α nuclear translocation in mesenchymal stem cells during hypoxia adaptation. Cell Death Differ 2018; 26:1716-1734. [PMID: 30464225 PMCID: PMC6748134 DOI: 10.1038/s41418-018-0241-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/11/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
Hypoxia inducible factor 1α (HIF1α) is a master regulator leading to metabolic adaptation, an essential physiological process to maintain the survival of stem cells under hypoxia. However, it is poorly understood how HIF1α translocates into the nucleus in stem cells under hypoxia. Here, we investigated the role of a motor adaptor protein Bicaudal D homolog 1 (BICD1) in dynein-mediated HIF1α nuclear translocation and the effect of BICD1 regulation on hypoxia adaptation and its therapeutic potential on human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs). In our results, silencing of BICD1 but not BICD2 abolished HIF1α nuclear translocation and its activity. BICD1 overexpression further enhanced hypoxia-induced HIF1α nuclear translocation. Hypoxia stimulated direct bindings of HIF1α to BICD1 and the intermediate chain of dynein (Dynein IC), which was abolished by BICD1 silencing. Akt inhibition reduced the binding of BICD1 to HIF1α and nuclear translocation of HIF1α. Conversely, Akt activation or GSK3β silencing further enhanced the hypoxia-induced HIF1α nuclear translocation. Furthermore, BICD1 silencing abolished hypoxia-induced glycolytic reprogramming and increased mitochondrial ROS accumulation and apoptosis in UCB-MSCs under hypoxia. In the mouse skin wound healing model, the transplanted cell survival and skin wound healing capacities of hypoxia-pretreated UCB-MSCs were reduced by BICD1 silencing and further increased by GSK3β silencing. In conclusion, we demonstrated that BICD1-induced HIF1α nuclear translocation is critical for hypoxia adaptation, which determines the regenerative potential of UCB-MSCs.
Collapse
|
529
|
Minassi A, Rogati F, Cruz C, Prados ME, Galera N, Jinénez C, Appendino G, Bellido ML, Calzado MA, Caprioglio D, Muñoz E. Triterpenoid Hydroxamates as HIF Prolyl Hydrolase Inhibitors. JOURNAL OF NATURAL PRODUCTS 2018; 81:2235-2243. [PMID: 30350996 DOI: 10.1021/acs.jnatprod.8b00514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Pentacyclic triterpenoid acids (PCTTAs) are pleiotropic agents that target many macromolecular end-points with low to moderate affinity. To explore the biological space associated with PCTTAs, we have investigated the carboxylate-to-hydroxamate transformation, discovering that it de-emphasizes affinity for the transcription factors targeted by the natural compounds (NF-κB, STAT3, Nrf2, TGR5) and selectively induces inhibitory activity on HIF prolyl hydrolases (PHDs). Activity was reversible, isoform-selective, dependent on the hydroxamate location, and negligible when this group was replaced by other chelating elements or O-alkylated. The hydroxamate of betulinic acid (5b) was selected for further studies, and evaluation of its effect on HIF-1α expression under normal and hypoxic conditions qualified it as a promising lead structure for the discovery of new candidates in the realm of neuroprotection.
Collapse
Affiliation(s)
- Alberto Minassi
- Dipartimento di Scienze del Farmaco , Università del Piemonte Orientale , Via Bovio 6 , Novara 28100 , Italy
| | - Federica Rogati
- Dipartimento di Scienze del Farmaco , Università del Piemonte Orientale , Via Bovio 6 , Novara 28100 , Italy
| | - Cristina Cruz
- VivaCell Biotechnology SL. Parque Científico Tecnológico , Rabanales 21. Edificio Centauro , 14014 Córdoba , Spain
| | - M Eugenia Prados
- VivaCell Biotechnology SL. Parque Científico Tecnológico , Rabanales 21. Edificio Centauro , 14014 Córdoba , Spain
| | - Nuria Galera
- VivaCell Biotechnology SL. Parque Científico Tecnológico , Rabanales 21. Edificio Centauro , 14014 Córdoba , Spain
| | - Carla Jinénez
- Maimonides Biomedical Research Institute of Córdoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- Department of Cellular Biology, Physiology and Immunology , University of Cordoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- University Hospital Reina Sofía , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
| | - Giovanni Appendino
- Dipartimento di Scienze del Farmaco , Università del Piemonte Orientale , Via Bovio 6 , Novara 28100 , Italy
| | - M Luz Bellido
- VivaCell Biotechnology SL. Parque Científico Tecnológico , Rabanales 21. Edificio Centauro , 14014 Córdoba , Spain
| | - Marco A Calzado
- Maimonides Biomedical Research Institute of Córdoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- Department of Cellular Biology, Physiology and Immunology , University of Cordoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- University Hospital Reina Sofía , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
| | - Diego Caprioglio
- Dipartimento di Scienze del Farmaco , Università del Piemonte Orientale , Via Bovio 6 , Novara 28100 , Italy
| | - Eduardo Muñoz
- Maimonides Biomedical Research Institute of Córdoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- Department of Cellular Biology, Physiology and Immunology , University of Cordoba , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
- University Hospital Reina Sofía , Avenida Menendez Pidal s/n , 14004 Cordoba , Spain
| |
Collapse
|
530
|
Onoue Y, Izumiya Y, Hanatani S, Ishida T, Arima Y, Yamamura S, Kimura Y, Araki S, Ishii M, Nakamura T, Oimatsu Y, Sakamoto K, Yamamoto E, Kojima S, Kaikita K, Tsujita K. Akt1-Mediated Muscle Growth Promotes Blood Flow Recovery After Hindlimb Ischemia by Enhancing Heme Oxygenase-1 in Neighboring Cells. Circ J 2018; 82:2905-2912. [DOI: 10.1253/circj.cj-18-0135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yoshiro Onoue
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Toshifumi Ishida
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Satoru Yamamura
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Yuichi Kimura
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Taishi Nakamura
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Yu Oimatsu
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Sunao Kojima
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University
| |
Collapse
|
531
|
Jin PP, Shao SY, Wu WT, Zhao XY, Huang BF, Fu QH, Que RS, Hu QD. Combination of transarterial chemoembolization and sorafenib improves outcomes of unresectable hepatocellular carcinoma: an updated systematic review and meta-analysis. Jpn J Clin Oncol 2018; 48:1058-1069. [PMID: 30272196 DOI: 10.1093/jjco/hyy138] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/18/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Piao-Piao Jin
- Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shi-Yi Shao
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wang-Teng Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Yu Zhao
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Feng Huang
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi-Han Fu
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ri-Sheng Que
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi-Da Hu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
532
|
Lai AG, Forde D, Chang WH, Yuan F, Zhuang X, Orbegozo Rubio C, Song CX, McKeating JA. Glucose and glutamine availability regulate HepG2 transcriptional responses to low oxygen. Wellcome Open Res 2018; 3:126. [PMID: 30345392 PMCID: PMC6178907 DOI: 10.12688/wellcomeopenres.14839.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2018] [Indexed: 01/04/2023] Open
Abstract
Background: Little is known about the impact of nutrients on cellular transcriptional responses, especially in face of environmental stressors such as oxygen deprivation. Hypoxia-inducible factors (HIF) coordinate the expression of genes essential for adaptation to oxygen-deprived environments. A second family of oxygen-sensing genes known as the alpha-ketoglutarate-dependent dioxygenases are also implicated in oxygen homeostasis and epigenetic regulation. The relationship between nutritional status and cellular response to hypoxia is understudied. In vitro cell culture systems frequently propagate cells in media that contains excess nutrients, and this may directly influence transcriptional response in hypoxia. Methods: We studied the effect of glucose and glutamine concentration on HepG2 hepatoma transcriptional response to low oxygen and expression of hypoxia inducible factor-1α (HIF-1α). Mass spectrometry confirmed low oxygen perturbation of dioxygenase transcripts resulted in changes in DNA methylation. Results: Under normoxic conditions, we observed a significant upregulation of both HIF-target genes and oxygen-dependent dioxygenases in HepG2 cells cultured with physiological levels of glucose or glutamine relative to regular DMEM media, demonstrating that excess glutamine/glucose can mask changes in gene expression. Under hypoxic conditions, CA9 was the most upregulated gene in physiological glutamine media while TETs and FTO dioxygenases were downregulated in physiological glucose. Hypoxic regulation of these transcripts did not associate with changes in HIF-1α protein expression. Downregulation of TETs suggests a potential for epigenetic modulation. Mass-spectrometry quantification of modified DNA bases confirmed our transcript data. Hypoxia resulted in decreased DNA hydroxymethylation, which correlated with TETs downregulation. Additionally, we observed that TET2 expression was significantly downregulated in patients with hepatocellular carcinoma, suggesting that tumour hypoxia may deregulate TET2 expression resulting in global changes in DNA hydroxymethylation. Conclusion: Given the dramatic effects of nutrient availability on gene expression, future in vitro experiments should be aware of how excess levels of glutamine and glucose may perturb transcriptional responses.
Collapse
Affiliation(s)
- Alvina G. Lai
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Donall Forde
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Wai Hoong Chang
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Fang Yuan
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, UK
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry, Peking University, Beijing, 100871, China
| | - Xiaodong Zhuang
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Claudia Orbegozo Rubio
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Chun-Xiao Song
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jane A. McKeating
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| |
Collapse
|
533
|
A feed-forward loop between nuclear translocation of CXCR4 and HIF-1α promotes renal cell carcinoma metastasis. Oncogene 2018; 38:881-895. [PMID: 30177838 PMCID: PMC6367212 DOI: 10.1038/s41388-018-0452-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/24/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) has been suggested to play a critical role in cancer metastasis. Some studies have described CXCR4 nuclear localization in metastatic lesions of renal cell carcinoma (RCC), which has been suggested to be correlated with cancer metastasis. However, the underlying mechanism and clinical significance of CXCR4 nuclear localization remains unknown. Here, we show that CXCR4 nuclear localization is more likely to occur in RCC tissues, especially in metastases, and is associated with poor prognosis. CXCR4 nuclear localization requires its nuclear localization sequence (NLS, residues 146-RPRK-149). After the mutation of NLS in CXCR4, CXCR4 nuclear localization in RCC cells is lost. Nuclear localization of CXCR4 promoted RCC tumorigenicity both in vitro and in vivo. Mechanistically, we found that CXCR4 and hypoxia-inducible factor-1α (HIF-1α) colocalized in RCC cells and interacted with each other. Moreover, CXCR4 nuclear localization promoted nuclear accumulation of HIF-1α, thereby promoting the expression of genes downstream of HIF-1α. Reciprocally, nuclear HIF-1α promoted CXCR4 transcription, thus forming a feed-forward loop. Subcellular CXCR4 and HIF-1α expression levels were independent adverse prognostic factors and could be combined with TNM stage to generate a predictive nomogram of the clinical outcome of patients with RCC. Therefore, our findings indicate that CXCR4 nuclear translocation plays a critical role in RCC metastasis and may serve as a prognostic biomarker and potential therapeutic target.
Collapse
|
534
|
Marsboom G, Rehman J. Hypoxia Signaling in Vascular Homeostasis. Physiology (Bethesda) 2018; 33:328-337. [PMID: 30109825 PMCID: PMC6230550 DOI: 10.1152/physiol.00018.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023] Open
Abstract
Hypoxia signaling in the vasculature controls vascular permeability, inflammation, vascular growth, and repair of vascular injury. In this review, we summarize recent insights in this burgeoning field and highlight the importance of studying the heterogeneity of hypoxia responses among individual patients, distinct vascular beds, and even individual vascular cells.
Collapse
Affiliation(s)
- Glenn Marsboom
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
- Department of Medicine, Section of Cardiology, University of Illinois College of Medicine , Chicago, Illinois
| |
Collapse
|
535
|
Araos J, Sleeman JP, Garvalov BK. The role of hypoxic signalling in metastasis: towards translating knowledge of basic biology into novel anti-tumour strategies. Clin Exp Metastasis 2018; 35:563-599. [DOI: 10.1007/s10585-018-9930-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023]
|
536
|
Zhou X, Liu C, Lu J, Zhu L, Li M. 2-Methoxyestradiol inhibits hypoxia-induced scleroderma fibroblast collagen synthesis by phosphatidylinositol 3-kinase/Akt/mTOR signalling. Rheumatology (Oxford) 2018; 57:1675-1684. [PMID: 29905853 DOI: 10.1093/rheumatology/key166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Indexed: 01/01/2023] Open
Abstract
Objectives To investigate the mechanism of 2-methoxyestradiol (2-ME) in inhibiting hypoxia-induced collagen synthesis of fibroblasts in SSc. Methods The expressions of hypoxia-inducible factor 1 alpha (HIF-1α) and connective tissue growth factor (CTGF) in skin specimens derived from SSc patients and healthy volunteers were examined by immunohistochemistry. HIF-1α was knocked down by lentiviral transduction, and SSc dermal fibroblasts cultured under normoxic (21% O2) or hypoxic (1% O2) condition were treated with PI3K inhibitor LY294002, rapamycin or 2-ME (25 μM). The protein levels of HIF-1α, CTGF, collagen I, p-Akt and p-mTOR were examined by western blotting or immunofluorescence. Apoptosis and cell cycle of fibroblasts were assessed by flow cytometry and by measuring caspase 3 activity, and cell proliferation was evaluated by Cell Counting Kit-8. Results The expressions of HIF-1α and CTGF were increased in skins of SSc patients compared with healthy controls. Hypoxia up-regulated the protein levels of HIF-1α, CTGF and collagen I in SSc fibroblasts. In contrast, 2-ME inhibited PI3K/Akt/mTOR pathway and down-regulated protein levels of HIF-1α, CTGF and collagen I. Knockdown of HIF-1α reduced expressions of CTGF and collagen I, which were further down-regulated by 2-ME intervention. Moreover, 2-ME promoted the apoptosis and inhibited the proliferation of SSc fibroblasts by arresting the cell cycle at the G2/M phase. Conclusion 2-ME reduced the production of CTGF and collagen I in SSc fibroblasts induced by hypoxia through PI3K/Akt/mTOR/HIF-1α signalling and inhibited the proliferation of fibroblasts. These findings suggested that 2-ME could be employed as a promising antifibrotic therapy for SSc.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chaofan Liu
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinghao Lu
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lubing Zhu
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
537
|
Abstract
Mitochondria are sensitive to numerous environmental stresses, which can lead to activation of mitochondrial stress responses (MSRs). Of particular recent interest has been the mitochondrial unfolded protein response (UPRmt), activated to restore protein homeostasis (proteostasis) upon mitochondrial protein misfolding. Several axes of the UPRmt have been described, creating some confusion as to the nature of the different responses. While distinct molecularly, these different axes are likely mutually beneficial and activated in parallel. This review aims at describing and distinguishing the different mammalian MSR/UPRmt axes to define key processes and members and to examine the involvement of protein misfolding.
Collapse
Affiliation(s)
- Christian Münch
- Institute of Biochemistry II, Goethe University - Medical Faculty, University Hospital, Frankfurt am Main, Germany.
| |
Collapse
|
538
|
Lu H, Tran L, Park Y, Chen I, Lan J, Xie Y, Semenza GL. Reciprocal Regulation of DUSP9 and DUSP16 Expression by HIF1 Controls ERK and p38 MAP Kinase Activity and Mediates Chemotherapy-Induced Breast Cancer Stem Cell Enrichment. Cancer Res 2018; 78:4191-4202. [PMID: 29880481 DOI: 10.1158/0008-5472.can-18-0270] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/08/2018] [Accepted: 06/01/2018] [Indexed: 02/05/2023]
Abstract
Triple-negative breast cancer (TNBC) has a poor prognosis due to its aggressive characteristics and lack of targeted therapies. Cytotoxic chemotherapy may reduce tumor bulk, but leaves residual disease due to the persistence of chemotherapy-resistant breast cancer stem cells (BCSC), which are critical for tumor recurrence and metastasis. Here, we demonstrate that hypoxia-inducible factor (HIF)-1-dependent regulation of mitogen-activated protein kinase (MAPK) signaling pathways contributes to chemotherapy-induced BCSC enrichment. Chemotherapy increased DUSP9 expression and decreased DUSP16 expression in a HIF1-dependent manner, leading to inhibition of ERK and activation of p38 signaling pathways, respectively. Inhibition of ERK caused transcriptional induction of the pluripotency factor Nanog through decreased inactivating phosphorylation of FoxO3, while activation of p38 stabilized Nanog and Klf4 mRNA through increased inactivating phosphorylation of RNA-binding protein ZFP36L1, both of which promoted specification of the BCSC phenotype. Inhibition of HIF1 or p38 signaling blocked chemotherapy-induced pluripotency factor expression and BCSC enrichment. These surprising results delineate a mechanism by which a transcription factor switches cells from ERK to p38 signaling in response to chemotherapy and suggest that therapeutic targeting of HIF1 or the p38 pathway in combination with chemotherapy will block BCSC enrichment and improve outcome in TNBC.Significance: These findings provide a molecular mechanism that may account for the increased relapse rate of women with TNBC who are treated with cytotoxic chemotherapy and suggest that combining chemotherapy with an inhibitor of HIF1 or p38 activity may increase patient survival. Cancer Res; 78(15); 4191-202. ©2018 AACR.
Collapse
Affiliation(s)
- Haiquan Lu
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Linh Tran
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngrok Park
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ivan Chen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jie Lan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yangyiran Xie
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Gregg L Semenza
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
539
|
García-Vásquez C, Fernández-Aceñero MJ, García Gómez-Heras S, Pastor C. Fibrin patch influences the expression of hypoxia-inducible factor-1α and nuclear factor-κBp65 factors on ischemic intestinal anastomosis. Exp Biol Med (Maywood) 2018; 243:803-808. [PMID: 29932372 PMCID: PMC6022912 DOI: 10.1177/1535370218777216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal anastomotic failure is one of the most severe complications in gastrointestinal surgery. Despite the great surgical improvements during the last decade, anastomotic leak rates remain practically the same, with a dramatically high grade of morbidity for patients. Leakages are usually the final consequence of ischemia in the anastomosis, leading to tissue hypoxia. In response to hypoxia, the cell orchestrates a variety of coordinated responses in order to restore oxygen homeostasis. The molecular mechanism of hypoxia sensitivity involves oxygen sensing hydroxylases, prolyl-hydroxylases, orchestrating two main transcription factors related to induction of inflammation and angiogenesis, namely nuclear factor-κB and hypoxia-inducible factors. The immunohistochemical expression of two transcription factors hypoxia-inducible factors-1α and nuclear factor-κB p65 has already been described in several disorders, including wound healing, asthma and chronic obstructive lung disease, rheumatoid arthritis, cancer, inflammatory bowel disease, and acute colitis. In the surgical field, fibrin sealants have been widely used to prevent leaks in lung surgery and they might also be useful as a reinforcement of sutures in intestinal anastomosis. The commercial fibrin sealant patches are hemostatic and adhesive surgical agents mainly derived from human plasma. We herein report the results of a prospective randomized experimental study on pigs. We performed a high-risk leakage model of bowel anastomosis, causing a significant devascularization of 10-15 cm of the bowel wall before performing a conventional colo-ileal anastomosis. We randomized the animals to receive a covering of the anastomosis with a fibrin patch (case group) or not (control group). We report the changes in the immunohistochemical expression of the proteins involved in tissue response to hypoxia in the experimental model. Our results indicate that the fibrin patch delays the healing response, promoting a longer lasting inflammation in the surgical bed. Nevertheless, the fibrin patches effectiveness to reduce dehiscence shown in clinical practice suggests that this delay does not negatively affect patients' outcome. Impact statement The consequences of the anastomotic failure are dramatic for patients. Understanding how the ever-increasing use of fibrin sealant, that seems to have a beneficial effect on the anastomoses, interacts with the tissue and the healing process can help to justify its use and encourage research on how to improve this effect even more. We feel that the present work shows that the patch can improve healing by complex mechanisms other than the mere contention and physical support of the intestine. Furthermore, research is needed to confirm our preliminary findings.
Collapse
|
540
|
Zhang J, Zhang Q. VHL and Hypoxia Signaling: Beyond HIF in Cancer. Biomedicines 2018; 6:biomedicines6010035. [PMID: 29562667 PMCID: PMC5874692 DOI: 10.3390/biomedicines6010035] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 12/12/2022] Open
Abstract
Von Hippel-Lindau (VHL) is an important tumor suppressor that is lost in the majority of clear cell carcinoma of renal cancer (ccRCC). Its regulatory pathway involves the activity of E3 ligase, which targets hypoxia inducible factor α (including HIF1α and HIF2α) for proteasome degradation. In recent years, emerging literature suggests that VHL also possesses other HIF-independent functions. This review will focus on VHL-mediated signaling pathways involving the latest identified substrates/binding partners, including N-Myc downstream-regulated gene 3 (NDRG3), AKT, and G9a, etc., and their physiological roles in hypoxia signaling and cancer. We will also discuss the crosstalk between VHL and NF-κB signaling. Lastly, we will review the latest findings on targeting VHL signaling in cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Qing Zhang
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
- Department of Pharmacology, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
541
|
Chen S, Chen X, Li W, Shan T, Lin WR, Ma J, Cui X, Yang W, Cao G, Li Y, Wang L, Kang Y. Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells. Oncol Lett 2018; 15:7144-7152. [PMID: 29731878 PMCID: PMC5921234 DOI: 10.3892/ol.2018.8219] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Tumor metastasis is accompanied by a two-stage process of epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). Currently, the exact mechanisms underlying EMT-MET conversion are unclear. In the present study, the mechanisms by which primary sites (hypoxic) and homing sites (normoxic or hyperoxic) participate in EMT-MET conversion were evaluated. Pancreatic cancer cells were grown under different oxygenation conditions. Cell morphology and epithelial (E)-cadherin and vimentin expression were examined. Transwell chambers were used to examine tumor invasiveness, and scratch assays were performed to examine cell migration. Reverse transcription-polymerase chain reaction and western blot analysis were used to quantitate the mRNA and protein expression of E-cadherin, vimentin, Snail and hypoxia-inducible factor (HIF)-1α. BxPc-3 and Panc-1 cells grown under hypoxic conditions demonstrated increased partial EMT, reduced E-cadherin expression, and increased vimentin expression, compared with cells grown under normoxic or hyperoxic conditions. Cells grown under hypoxic conditions also indicated increased migration and invasiveness. HIF-1α mRNA and protein expression was increased in cells grown under hypoxic conditions. These changes were reversed when a specific inhibitor of the HIF-1α receptor was used to block HIF-1α signaling. Differences in oxygen concentration at primary sites and homing sites are important in the EMT-MET process, and the underlying mechanism may involve HIF-1α-Snail signaling.
Collapse
Affiliation(s)
- Shuo Chen
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xi Chen
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wei Li
- The Institute for Population and Development Studies, School of Public Policy and Administration, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Tao Shan
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wan Run Lin
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Jiancang Ma
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xijuan Cui
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenbin Yang
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Gang Cao
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yiming Li
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Li Wang
- Department of Gastrointestinal Surgery, Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| | - Ya'an Kang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
542
|
Effects of an Acute Exercise Bout on Serum Hepcidin Levels. Nutrients 2018; 10:nu10020209. [PMID: 29443922 PMCID: PMC5852785 DOI: 10.3390/nu10020209] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 01/30/2018] [Accepted: 02/11/2018] [Indexed: 02/07/2023] Open
Abstract
Iron deficiency is a frequent and multifactorial disorder in the career of athletes, particularly in females. Exercise-induced disturbances in iron homeostasis produce deleterious effects on performance and adaptation to training; thus, the identification of strategies that restore or maintain iron homeostasis in athletes is required. Hepcidin is a liver-derived hormone that degrades the ferroportin transport channel, thus reducing the ability of macrophages to recycle damaged iron, and decreasing iron availability. Although it has been suggested that the circulating fraction of hepcidin increases during early post-exercise recovery (~3 h), it remains unknown how an acute exercise bout may modify the circulating expression of hepcidin. Therefore, the current review aims to determine the post-exercise expression of serum hepcidin in response to a single session of exercise. The review was carried out in the Dialnet, Elsevier, Medline, Pubmed, Scielo and SPORTDiscus databases, using hepcidin (and “exercise” or “sport” or “physical activity”) as a strategy of search. A total of 19 articles were included in the review after the application of the inclusion/exclusion criteria. This search found that a single session of endurance exercise (intervallic or continuous) at moderate or vigorous intensity (60–90% VO2peak) stimulates an increase in the circulating levels of hepcidin between 0 h and 6 h after the end of the exercise bout, peaking at ~3 h post-exercise. The magnitude of the response of hepcidin to exercise seems to be dependent on the pre-exercise status of iron (ferritin) and inflammation (IL-6). Moreover, oxygen disturbances and the activation of a hypoxia-induced factor during or after exercise may stimulate a reduction of hepcidin expression. Meanwhile, cranberry flavonoids supplementation promotes an anti-oxidant effect that may facilitate the post-exercise expression of hepcidin. Further studies are required to explore the effect of resistance exercise on hepcidin expression.
Collapse
|
543
|
Cerychova R, Pavlinkova G. HIF-1, Metabolism, and Diabetes in the Embryonic and Adult Heart. Front Endocrinol (Lausanne) 2018; 9:460. [PMID: 30158902 PMCID: PMC6104135 DOI: 10.3389/fendo.2018.00460] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
The heart is able to metabolize any substrate, depending on its availability, to satisfy its energy requirements. Under normal physiological conditions, about 95% of ATP is produced by oxidative phosphorylation and the rest by glycolysis. Cardiac metabolism undergoes reprograming in response to a variety of physiological and pathophysiological conditions. Hypoxia-inducible factor 1 (HIF-1) mediates the metabolic adaptation to hypoxia and ischemia, including the transition from oxidative to glycolytic metabolism. During embryonic development, HIF-1 protects the embryo from intrauterine hypoxia, its deletion as well as its forced expression are embryonically lethal. A decrease in HIF-1 activity is crucial during perinatal remodeling when the heart switches from anaerobic to aerobic metabolism. In the adult heart, HIF-1 protects against hypoxia, although its deletion in cardiomyocytes affects heart function even under normoxic conditions. Diabetes impairs HIF-1 activation and thus, compromises HIF-1 mediated responses under oxygen-limited conditions. Compromised HIF-1 signaling may contribute to the teratogenicity of maternal diabetes and diabetic cardiomyopathy in adults. In this review, we discuss the function of HIF-1 in the heart throughout development into adulthood, as well as the deregulation of HIF-1 signaling in diabetes and its effects on the embryonic and adult heart.
Collapse
Affiliation(s)
- Radka Cerychova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- *Correspondence: Gabriela Pavlinkova
| |
Collapse
|