1
|
Slawski J, Jaśkiewicz M, Barton A, Kozioł S, Collawn JF, Bartoszewski R. Regulation of the HIF switch in human endothelial and cancer cells. Eur J Cell Biol 2024; 103:151386. [PMID: 38262137 DOI: 10.1016/j.ejcb.2024.151386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/25/2024] Open
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors that reprogram the transcriptome for cells to survive hypoxic insults and oxidative stress. They are important during embryonic development and reprogram the cells to utilize glycolysis when the oxygen levels are extremely low. This metabolic change facilitates normal cell survival as well as cancer cell survival. The key feature in survival is the transition between acute hypoxia and chronic hypoxia, and this is regulated by the transition between HIF-1 expression and HIF-2/HIF-3 expression. This transition is observed in many human cancers and endothelial cells and referred to as the HIF Switch. Here we discuss the mechanisms involved in the HIF Switch in human endothelial and cancer cells which include mRNA and protein levels of the alpha chains of the HIFs. A major continuing effort in this field is directed towards determining the differences between normal and tumor cell utilization of this important pathway, and how this could lead to potential therapeutic approaches.
Collapse
Affiliation(s)
- Jakub Slawski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Maciej Jaśkiewicz
- International Research Agenda 3P, Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Anna Barton
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Sylwia Kozioł
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
2
|
Chowdhury M, Das PK. Hypoxia: Intriguing Feature in Cancer Cell Biology. ChemMedChem 2024; 19:e202300551. [PMID: 38328976 DOI: 10.1002/cmdc.202300551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
Hypoxia, a key aspect of the tumor microenvironment, plays a vital role in cell proliferation, angiogenesis, metabolism, and the immune response within tumors. These factors collectively promote tumor advancement, aggressiveness, metastasis and result in a poor prognosis. Hypoxia inducible factor 1α (HIF-1α), activated under low oxygen conditions, mediates many of these effects by altering drug target expression, metabolic regulation, and oxygen consumption. These changes promote cancer cell growth and survival. Hypoxic tumor cells develop aggressive traits and resistance to chemotherapy and radiotherapy, leading to increased mortality. Targeting hypoxic tumor offers a potential solution to overcome the challenges posed by tumor heterogeneity and can be used in designing diagnostic and therapeutic nanocarriers for various solid cancers. This concept provides an overview of the intricate relationship between hypoxia and the tumor microenvironment, highlighting its potential as a promising tool for cancer therapies. The article explores the development of hypoxia in cancer cells and its role in cancer progression, along with the latest advancements in hypoxia-triggered cancer treatment.
Collapse
Affiliation(s)
- Monalisa Chowdhury
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India
| |
Collapse
|
3
|
Elbassiouny AA, Buck LT, Abatti LE, Mitchell JA, Crampton WGR, Lovejoy NR, Chang BSW. Evolution of a novel regulatory mechanism of hypoxia inducible factor in hypoxia-tolerant electric fishes. J Biol Chem 2024; 300:105727. [PMID: 38325739 PMCID: PMC10958119 DOI: 10.1016/j.jbc.2024.105727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Hypoxia is a significant source of metabolic stress that activates many cellular pathways involved in cellular differentiation, proliferation, and cell death. Hypoxia is also a major component in many human diseases and a known driver of many cancers. Despite the challenges posed by hypoxia, there are animals that display impressive capacity to withstand lethal levels of hypoxia for prolonged periods of time and thus offer a gateway to a more comprehensive understanding of the hypoxic response in vertebrates. The weakly electric fish genus Brachyhypopomus inhabits some of the most challenging aquatic ecosystems in the world, with some species experiencing seasonal anoxia, thus providing a unique system to study the cellular and molecular mechanisms of hypoxia tolerance. In this study, we use closely related species of Brachyhypopomus that display a range of hypoxia tolerances to probe for the underlying molecular mechanisms via hypoxia inducible factors (HIFs)-transcription factors known to coordinate the cellular response to hypoxia in vertebrates. We find that HIF1⍺ from hypoxia tolerant Brachyhypopomus species displays higher transactivation in response to hypoxia than that of intolerant species, when overexpressed in live cells. Moreover, we identified two SUMO-interacting motifs near the oxygen-dependent degradation and transactivation domains of the HIF1⍺ protein that appear to boost transactivation of HIF1, regardless of the genetic background. Together with computational analyses of selection, this shows that evolution of HIF1⍺ are likely to underlie adaptations to hypoxia tolerance in Brachyhypopomus electric fishes, with changes in two SUMO-interacting motifs facilitating the mechanism of this tolerance.
Collapse
Affiliation(s)
- Ahmed A Elbassiouny
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada.
| | - Leslie T Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Luis E Abatti
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | | | - Nathan R Lovejoy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada; Department of Biological Sciences, University of Toronto Scarborough, Scarborough, Ontario, Canada
| | - Belinda S W Chang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Ren Y, Xi Q, He Z, Sun H, Li S. Expression and Variations in EPO Associated with Oxygen Metabolism in Tibetan Sheep. Animals (Basel) 2024; 14:535. [PMID: 38396503 PMCID: PMC10886301 DOI: 10.3390/ani14040535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
After a long period of adaptive evolution, Tibetan sheep have adapted to the plateau environment in terms of genetics, physiology and biochemistry, but the mechanism of hypoxia adaptation has not been fully elucidated, and the functional genes and molecular mechanisms regulating the hypoxia adaptation of Tibetan sheep need to be further studied. In this study, Tibetan sheep were selected as the research object, and the mRNA expression levels of the hypoxa-related gene EPO in heart, lung, kidney, liver, spleen and longissimus dorsi muscle of Hu sheep (100 m) and Tibetan sheep at different altitudes (2500 m, 3500 m, 4500 m) were assessed by RT-qPCR. The SNPs loci were detected by sequencing and Kompetitive Allele-Specific PCR (KASP) technology, then the correlation between genetic polymorphism and blood gas was analyzed. The results show that the expression of the EPO gene was the highest in the kidney, indicating that the expression of EPO gene had tissue differences. The expression levels of the EPO gene in the heart, lung and liver of Tibetan sheep at a 4500 m altitude were significantly higher than those in Hu sheep (p < 0.05), and the levels in the hearts of Tibetan sheep increased with the increase in altitude. Three mutations were identified in the EPO gene, the SNPs (g.855 A > C) in exon 1 and the SNPs (g.1985 T > G and g.2115 G > C) in exon 4, which were named EPO-SNP1, EPO-SNP2 and EPO-SNP3, respectively, and all three SNPs showed three genotypes. Correlation analysis showed that g.2115 G > C sites were significantly correlated with pO2 (p < 0.05), and haplotype combinations were significantly correlated with pO2 (p < 0.05). Thesee results suggest that the expression of the EPO gene is altitude-differentiated and organ-differentiated, and the EPO gene variants have significant effects on pO2, which may be beneficial to the adaptation of Tibetan sheep to hypoxia stress.
Collapse
Affiliation(s)
- Yue Ren
- Institute of Livestock Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China
| | - Qiming Xi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhaohua He
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Hongxian Sun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shaobin Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
5
|
Gao X, Zhang Y, Zhu Q, Han Y, Jia R, Zhang W. Effects of myeloperoxidase on inflammatory responses with hypoxia in Citrobacter rodentium-infectious mice. Immun Inflamm Dis 2024; 12:e1157. [PMID: 38415976 PMCID: PMC10836036 DOI: 10.1002/iid3.1157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 02/29/2024] Open
Abstract
PURPOSE Myeloperoxidase (MPO) has been identified as a mediator in various inflammatory diseases. Bacterial infection of the intestine and hypoxia can both lead to inflammatory responses, but the role of MPO in these phenomena remains unclear. METHODS By building the MPO-/- mice, we evaluated relevant inflammatory factors and tissue damage in mice with intestinal Citrobacter rodentium infection and hypoxia. The body weight and excreted microorganisms were monitored. Intestinal tissues were collected 7 days after bacterial infection under hypoxia to undergo haematoxylin-eosin staining and assess the degree of pathological damage. ELISA assays were performed to quantify the serum levels of TNF-α, IFN-γ, IL-6, and IL-1β inflammatory cytokines. PCR, WB, and IF assays were conducted to determine the expression of chemokines MCP1, MIP2, and KC in the colon and spleen. RESULTS The C. rodentium infection and hypoxia caused weight loss, intestinal colitis, and splenic inflammatory cells active proliferation in wild-type mice. MPO deficiency alleviated this phenomenon. MPO-/- mice also displayed a significant decline in bacteria clearing ability. The level of TNF-α in the serum and spleen was both lower in MPO-/- hypoxia C. rodentium-infected mice than that in wild-type mice. The chemokines expression levels of MIP2, KC, and MCP1 in the spleen and colon of each bacterial infected group were significantly increased (p < .05), while in hypoxia, the factors in the spleen and colon were decreased (p < .05). MPO deficiency was found to lower the levels of these chemokines compared with wild-type mice. CONCLUSION MPO plays an important role of the inflammatory responses in infectious enteritis and hypoxia in mice, and the loss of MPO may greatly reduce the body's inflammatory responses to fight diseases.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Yu Zhang
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
| | - Qinfang Zhu
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Ying Han
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Ruhan Jia
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Wei Zhang
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| |
Collapse
|
6
|
Liu J, Jiang Y, Chen L, Qian Z, Zhang Y. Associations between HIFs and tumor immune checkpoints: mechanism and therapy. Discov Oncol 2024; 15:2. [PMID: 38165484 PMCID: PMC10761656 DOI: 10.1007/s12672-023-00836-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Hypoxia, which activates a variety of signaling pathways to enhance tumor cell growth and metabolism, is among the primary features of tumor cells. Hypoxia-inducible factors (HIFs) have a substantial impact on a variety of facets of tumor biology, such as epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and improved radiation resistance. HIFs induce hypoxia-adaptive responses in tumor cells. Many academics have presented preclinical and clinical research targeting HIFs in tumor therapy, highlighting the potential applicability of targeted HIFs. In recent years, the discovery of numerous pharmacological drugs targeting the regulatory mechanisms of HIFs has garnered substantial attention. Additionally, HIF inhibitors have attained positive results when used in conjunction with traditional oncology radiation and/or chemotherapy, as well as with the very promising addition of tumor immunotherapy. Immune checkpoint inhibitors (CPIs), which are employed in a range of cancer treatments over the past decades, are essential in tumor immunotherapy. Nevertheless, the use of immunotherapy has been severely hampered by tumor resistance and treatment-related toxicity. According to research, HIF inhibitors paired with CPIs may be game changers for multiple malignancies, decreasing malignant cell plasticity and cancer therapy resistance, among other things, and opening up substantial new pathways for immunotherapy drug development. The structure, activation mechanisms, and pharmacological sites of action of the HIF family are briefly reviewed in this work. This review further explores the interactions between HIF inhibitors and other tumor immunotherapy components and covers the potential clinical use of HIF inhibitors in combination with CPIs.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China.
| |
Collapse
|
7
|
Lian M, Mortoglou M, Uysal-Onganer P. Impact of Hypoxia-Induced miR-210 on Pancreatic Cancer. Curr Issues Mol Biol 2023; 45:9778-9792. [PMID: 38132457 PMCID: PMC10742176 DOI: 10.3390/cimb45120611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Pancreatic cancer (PC) poses significant clinical challenges, with late-stage diagnosis and limited therapeutic options contributing to its dismal prognosis. A hallmark feature of PC is the presence of a profoundly hypoxic tumour microenvironment, resulting from various factors such as fibrotic stroma, rapid tumour cell proliferation, and poor vascularization. Hypoxia plays a crucial role in promoting aggressive cancer behaviour, therapeutic resistance, and immunosuppression. Previous studies have explored the molecular mechanisms behind hypoxia-induced changes in PC, focusing on the role of hypoxia-inducible factors (HIFs). Among the myriad of molecules affected by hypoxia, microRNA-210 (miR-210) emerges as a central player. It is highly responsive to hypoxia and regulated by HIF-dependent and HIF-independent pathways. miR-210 influences critical cellular processes, including angiogenesis, metastasis, and apoptosis, all of which contribute to PC progression and resistance to treatment. Understanding these pathways provides insights into potential therapeutic targets. Furthermore, investigating the role of miR-210 and its regulation in hypoxia sheds light on the potential development of early diagnostic strategies, which are urgently needed to improve outcomes for PC patients. This review delves into the complexities of PC and introduces the roles of hypoxia and miR-210 in the progression of PC.
Collapse
Affiliation(s)
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK; (M.L.); (M.M.)
| |
Collapse
|
8
|
Alsharabasy AM, Aljaabary A, Bohara R, Farràs P, Glynn SA, Pandit A. Nitric Oxide-Scavenging, Anti-Migration Effects, and Glycosylation Changes after Hemin Treatment of Human Triple-Negative Breast Cancer Cells: A Mechanistic Study. ACS Pharmacol Transl Sci 2023; 6:1416-1432. [PMID: 37854626 PMCID: PMC10580390 DOI: 10.1021/acsptsci.3c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Indexed: 10/20/2023]
Abstract
The enhanced expression of nitric oxide (•NO) synthase predicts triple-negative breast cancer outcome and its resistance to different therapeutics. Our earlier work demonstrated the efficiency of hemin to scavenge the intra- and extracellular •NO, proposing its potency as a therapeutic agent for inhibiting cancer cell migration. In continuation, the present work evaluates the effects of •NO on the migration of MDA-MB-231 cells and how hemin modulates the accompanied cellular behavior, focusing on the corresponding expression of cellular glycoproteins, migration-associated markers, and mitochondrial functions. We demonstrated for the first time that while •NO induced cell migration, hemin contradicted that by •NO-scavenging. This was in combination with modulation of the •NO-enhanced glycosylation patterns of cellular proteins with inhibition of the expression of specific proteins involved in the epithelial-mesenchymal transition. These effects were in conjunction with changes in the mitochondrial functions related to both •NO, hemin, and its nitrosylated product. Together, these results suggest that hemin can be employed as a potential anti-migrating agent targeting •NO-scavenging and regulating the expression of migration-associated proteins.
Collapse
Affiliation(s)
- Amir M. Alsharabasy
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Amal Aljaabary
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Raghvendra Bohara
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Pau Farràs
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- School
of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway H91 TK33, Ireland
| | - Sharon A. Glynn
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- Discipline
of Pathology, Lambe Institute for Translational Research, School of
Medicine, University of Galway, Galway H91 YR71, Ireland
| | - Abhay Pandit
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
9
|
Huang Q, Qiao Lv, Jiang L, Chen Q, Zhang K. Recent progress of biocompatible carbon dots in hypoxia-related fields. J Biomater Appl 2023; 37:1159-1168. [PMID: 36083209 DOI: 10.1177/08853282221125313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Almost all eukaryotes need oxygen to maintain regular physiological activities. When the organism is under hypoxic situation for a persistent or periodic, it will induce irreversible physiological disorders and even pathological results. Hypoxia is closely related to the pathogenesis of metabolic diseases, cancer, chronic heart disease and kidney disease, myocardial ischemia, as well as reproductive diseases like preeclampsia and endometriosis. Therefore, monitoring and treatment of hypoxia have important implications for the pathophysiology of human-related diseases. Carbon dots (CDs) are emerging nanomaterials developed after 2004 with excellent performance, and have broad application potential in variousdomains likeoptical, biomedicine, energy. Advanced hypoxia therapeutics should be integrated with monitoring and treatment, and CDs with excellent performance are good potential options when sensing is combined with various therapeutic methods. Some researchers have also begun to carry out research in related fields and achieved some results. This article aims to clarify the various applications of CDs in hypoxia-related fields in recent years, including hypoxia sensing and hypoxia tumor theranostics. Finally, the possible challenges and prospects for the application of CDs in hypoxia-related fields are discussed.
Collapse
Affiliation(s)
- Qing Huang
- Clinical Medicine Research Center, Xinqiao Hospital, 12525Army Medical UniversityThird Military Medical University, Chongqing, China
| | - Qiao Lv
- Clinical Medicine Research Center, Xinqiao Hospital, 12525Army Medical UniversityThird Military Medical University, Chongqing, China
| | - Lu Jiang
- Clinical Medicine Research Center, Xinqiao Hospital, 12525Army Medical UniversityThird Military Medical University, Chongqing, China
| | - Qian Chen
- Clinical Medicine Research Center, Xinqiao Hospital, 12525Army Medical UniversityThird Military Medical University, Chongqing, China
| | - Kebin Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, 12525Army Medical UniversityThird Military Medical University, Chongqing, China
| |
Collapse
|
10
|
Körbelin J, Klein J, Matuszcak C, Runge J, Harbaum L, Klose H, Hennigs JK. Transcription factors in the pathogenesis of pulmonary arterial hypertension-Current knowledge and therapeutic potential. Front Cardiovasc Med 2023; 9:1036096. [PMID: 36684555 PMCID: PMC9853303 DOI: 10.3389/fcvm.2022.1036096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/21/2022] [Indexed: 01/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by elevated pulmonary vascular resistance and pulmonary artery pressure. Mortality remains high in severe cases despite significant advances in management and pharmacotherapy. Since currently approved PAH therapies are unable to significantly reverse pathological vessel remodeling, novel disease-modifying, targeted therapeutics are needed. Pathogenetically, PAH is characterized by vessel wall cell dysfunction with consecutive remodeling of the pulmonary vasculature and the right heart. Transcription factors (TFs) regulate the process of transcribing DNA into RNA and, in the pulmonary circulation, control the response of pulmonary vascular cells to macro- and microenvironmental stimuli. Often, TFs form complex protein interaction networks with other TFs or co-factors to allow for fine-tuning of gene expression. Therefore, identification of the underlying molecular mechanisms of TF (dys-)function is essential to develop tailored modulation strategies in PAH. This current review provides a compendium-style overview of TFs and TF complexes associated with PAH pathogenesis and highlights their potential as targets for vasculoregenerative or reverse remodeling therapies.
Collapse
Affiliation(s)
- Jakob Körbelin
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Jakob Körbelin,
| | - Julius Klein
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane Matuszcak
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Runge
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Harbaum
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Klose
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan K. Hennigs
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Jan K. Hennigs,
| |
Collapse
|
11
|
Jaśkiewicz M, Moszyńska A, Króliczewski J, Cabaj A, Bartoszewska S, Charzyńska A, Gebert M, Dąbrowski M, Collawn JF, Bartoszewski R. The transition from HIF-1 to HIF-2 during prolonged hypoxia results from reactivation of PHDs and HIF1A mRNA instability. Cell Mol Biol Lett 2022; 27:109. [PMID: 36482296 PMCID: PMC9730601 DOI: 10.1186/s11658-022-00408-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
The hypoxia-inducible factors (HIF) are transcription factors that activate the adaptive hypoxic response when oxygen levels are low. The HIF transcriptional program increases oxygen delivery by inducing angiogenesis and by promoting metabolic reprograming that favors glycolysis. The two major HIFs, HIF-1 and HIF-2, mediate this response during prolonged hypoxia in an overlapping and sequential fashion that is referred to as the HIF switch. Both HIF proteins consist of an unstable alpha chain and a stable beta chain. The instability of the alpha chains is mediated by prolyl hydroxylase (PHD) activity during normoxic conditions, which leads to ubiquitination and proteasomal degradation of the alpha chains. During normoxic conditions, very little HIF-1 or HIF-2 alpha-beta dimers are present because of PHD activity. During hypoxia, however, PHD activity is suppressed, and HIF dimers are stable. Here we demonstrate that HIF-1 expression is maximal after 4 h of hypoxia in primary endothelial cells and then is dramatically reduced by 8 h. In contrast, HIF-2 is maximal at 8 h and remains elevated up to 24 h. There are differences in the HIF-1 and HIF-2 transcriptional profiles, and therefore understanding how the transition between them occurs is important and not clearly understood. Here we demonstrate that the HIF-1 to HIF-2 transition during prolonged hypoxia is mediated by two mechanisms: (1) the HIF-1 driven increase in the glycolytic pathways that reactivates PHD activity and (2) the much less stable mRNA levels of HIF-1α (HIF1A) compared to HIF-2α (EPAS1) mRNA. We also demonstrate that the alpha mRNA levels directly correlate to the relative alpha protein levels, and therefore to the more stable HIF-2 expression during prolonged hypoxia.
Collapse
Affiliation(s)
- Maciej Jaśkiewicz
- grid.11451.300000 0001 0531 3426International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Adrianna Moszyńska
- grid.11451.300000 0001 0531 3426Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Jarosław Króliczewski
- grid.11451.300000 0001 0531 3426Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Aleksandra Cabaj
- grid.419305.a0000 0001 1943 2944Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Sylwia Bartoszewska
- grid.11451.300000 0001 0531 3426Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Agata Charzyńska
- grid.419305.a0000 0001 1943 2944Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Magda Gebert
- grid.11451.300000 0001 0531 3426Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Michał Dąbrowski
- grid.419305.a0000 0001 1943 2944Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - James F. Collawn
- grid.265892.20000000106344187Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, BirminghamBirmingham, AL 35233 USA
| | - Rafal Bartoszewski
- grid.8505.80000 0001 1010 5103Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| |
Collapse
|
12
|
Zhou D, Wang C, Zheng J, Zhao J, Wei S, Xiong Y, Limbu SM, Kong Y, Cao F, Ding Z. Dietary thiamine modulates carbohydrate metabolism, antioxidant status, and alleviates hypoxia stress in oriental river prawn Macrobrachium nipponense (de Haan). FISH & SHELLFISH IMMUNOLOGY 2022; 131:42-53. [PMID: 36191902 DOI: 10.1016/j.fsi.2022.09.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 06/16/2023]
Abstract
Hypoxia is one of the challenges in prawns aquaculture. However, the role of thiamine, which is a coenzyme in carbohydrate metabolism with antioxidant properties, in reducing hypoxia in prawns aquaculture is currently unknown. We investigated the effects of thiamine on antioxidant status, carbohydrate metabolism and acute hypoxia in oriental river prawn, Macrobrachium nipponense. One thousand eight hundred prawns (0.123 ± 0.003 g) were fed five diets (60 prawns each tank, six replicates per diet) supplemented with graded thiamine levels (5.69, 70.70, 133.67, 268.33 and 532.00 mg/kg dry mater) for eight weeks and then exposed to hypoxia stress for 12 h followed by reoxyegnation for 12 h. The results showed that, under normoxia, prawns fed the 133.67 or 268.33 mg/kg thiamine diet had significantly lower glucose 6-phosphatedehydrogenase, succinate dehydrogenase and phosphoenolpyruvate carboxykinase activities than those fed the other diets. Moreover, total antioxidant capacity (T-AOC) increased significantly when prawns were fed the 133.67 mg/kg thiamine diet. Superoxide dismutase (SOD) activity and malonaldehyde (MDA) content also increased significantly when prawns were fed the 268.33 or 532.00 mg/kg thiamine diet under hypoxia. And the significantly increased SOD activity and MDA level also observed in prawns fed 532.00 mg/kg thiamine under reoxygenation. Under normoxia, prawns fed the 70.70 or 133.67 mg/kg thiamine diet decreased the mRNA expressions of AMP-activated protein kinase-alpha (AMPK-α), pyruvate dehydrogenase-E1-α subunit (PDH-E1-α) and hypoxia-inducible factor-1s (HIF-1α, HIF-1β), but increased the mRNA expressions of phosphofructokinase (PFK) significantly. After 12 h of hypoxia, the energy metabolism related genes (AMPK-β, AMPK-γ, PFK, PDH-E1-α), hypoxia-inducible factor related genes (HIF-1α, HIF-1β) and thiamine transporter gene (SLC19A2) were up-regulated significantly in prawns fed the 133.67 or 268.33 mg/kg thiamine diets. After 12 h of reoxygenation, prawns fed the 133.67 or 268.33 mg/kg diet significantly decreased the SOD activity, MDA level and SLC19A2 mRNA expression compared with other diets. The optimum thiamine was 161.20 mg/kg for minimum MDA content and 143.17 mg/kg for maximum T-AOC activity based on cubic regression analysis. In summary, supplementing 143.17 to 161.20 mg/kg thiamine in the diets for M. nipponense improves the antioxidant capacity under normoxia and reduces the oxidative damage under hypoxia stress.
Collapse
Affiliation(s)
- Dongsheng Zhou
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Chengli Wang
- Jiangsu Agri-animal Husbandry Vocational College, Jiangsu, China
| | - Jinxian Zheng
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Jianhua Zhao
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Shanshan Wei
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Yunfeng Xiong
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Samwel Mchele Limbu
- Department of Aquaculture Technology, School of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, P.O. Box 35091, Dar es Salaam, Tanzania
| | - Youqin Kong
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Fang Cao
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Zhili Ding
- Zhejiang Provincial Key Laboratory of Aquatic Resources Conservation and Development, College of Life Science, Huzhou University, Huzhou, Zhejiang, 313000, China.
| |
Collapse
|
13
|
Moszyńska A, Jaśkiewicz M, Serocki M, Cabaj A, Crossman DK, Bartoszewska S, Gebert M, Dąbrowski M, Collawn JF, Bartoszewski R. The hypoxia-induced changes in miRNA-mRNA in RNA-induced silencing complexes and HIF-2 induced miRNAs in human endothelial cells. FASEB J 2022; 36:e22412. [PMID: 35713587 DOI: 10.1096/fj.202101987r] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/11/2022]
Abstract
The cellular adaptive response to hypoxia relies on the expression of hypoxia-inducible factors (HIFs), HIF-1 and HIF-2. HIFs regulate global gene expression changes during hypoxia that are necessary for restoring oxygen homeostasis and promoting cell survival. In the early stages of hypoxia, HIF-1 is elevated, whereas at the later stages, HIF-2 becomes the predominant form. What governs the transition between the two HIFs (the HIF switch) and the role of miRNAs in this regulation are not completely clear. Genome-wide expression studies on the miRNA content of RNA-induced silencing complexes (RISC) in HUVECs exposed to hypoxia compared to the global miRNA-Seq analysis revealed very specific differences between these two populations. We analyzed the miRNA and mRNA composition of RISC at 2 h (mainly HIF-1 driven), 8 h (HIF-1 and HIF-2 elevated), and 16 h (mainly HIF-2 driven) in a gene ontology context. This allowed for determining the direct impact of the miRNAs in modulating the cellular signaling pathways involved in the hypoxic adaptive response. Our results indicate that the miRNA-mRNA RISC components control the adaptive responses, and this does not always rely on the miRNA transcriptional elevations during hypoxia. Furthermore, we demonstrate that the hypoxic levels of the vast majority of HIF-1-dependent miRNAs (including miR-210-3p) are also HIF-2 dependent and that HIF-2 governs the expression of 11 specific miRNAs. In summary, the switch from HIF-1 to HIF-2 during hypoxia provides an important level of miRNA-driven control in the adaptive pathways in endothelial cells.
Collapse
Affiliation(s)
- Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Maciej Jaśkiewicz
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Serocki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Aleksandra Cabaj
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - David K Crossman
- Department of Genetics, The UAB Genomics Core Facility, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
14
|
Effects of Prolonged Exposure to Hypobaric Hypoxia on Oxidative Stress: Overwintering in Antarctic Concordia Station. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4430032. [PMID: 35535360 PMCID: PMC9078816 DOI: 10.1155/2022/4430032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/25/2022] [Accepted: 04/09/2022] [Indexed: 12/14/2022]
Abstract
Concordia Station is the permanent, research station on the Antarctic Plateau at 3230 m. During the eleventh winter-over campaign (DC11-2015; February 2015 to November 2015) at Antarctic Concordia Station, 13 healthy team members were studied and blood samples were collected at six different time points: baseline measurements (T0), performed at sea level before the departure, and during the campaign at 3, 7, 20, 90, and 300 days after arrival at Concordia Station. Reducing the partial pressure of O2 as barometric pressure falls, hypobaric hypoxia (HH) triggers several physiological adaptations. Among the others, increased oxidative stress and enhanced generation of reactive oxygen/nitrogen species (ROS/RNS), resulting in severe oxidative damage, were observed, which can share potential physiopathological mechanisms associated with many diseases. This study characterized the extent and time-course changes after acute and chronic HH exposure, elucidating possible fundamental mechanisms of adaptation. ROS, oxidative stress biomarkers, nitric oxide, and proinflammatory cytokines significantly increased (range 24-135%) during acute and chronic hypoxia exposure (peak 20th day) with a decrease in antioxidant capacity (peak 90th day: -52%). Results suggest that the adaptive response of oxidative stress balance to HH requires a relatively long time, more than 300th days, as all the observed variables do not return to the preexposition level. These findings may also be relevant to patients in whom oxygen availability is limited through disease (i.e., chronic heart and lung and/or kidney disease) and/or during long-duration space missions.
Collapse
|
15
|
Breakthrough Science: Hypoxia-Inducible Factors, Oxygen Sensing, and Disorders of Hematopoiesis. Blood 2021; 139:2441-2449. [PMID: 34411243 DOI: 10.1182/blood.2021011043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/28/2021] [Indexed: 11/20/2022] Open
Abstract
Hypoxia-inducible factors (HIF) were discovered as activators of erythropoietin gene transcription in response to reduced O2 availability. O2-dependent hydroxylation of HIFs on proline and asparagine residues regulates protein stability and transcription activity, respectively. Mutations in genes encoding components of the oxygen sensing pathway cause familial erythrocytosis. Several small molecule inhibitors of HIF prolyl hydroxylases are currently in clinical trials as erythropoiesis stimulating agents. HIFs are overexpressed in bone marrow neoplasms, and the development of HIF inhibitors may improve outcome in these disorders.
Collapse
|
16
|
Casillas AL, Chauhan SS, Toth RK, Sainz AG, Clements AN, Jensen CC, Langlais PR, Miranti CK, Cress AE, Warfel NA. Direct phosphorylation and stabilization of HIF-1α by PIM1 kinase drives angiogenesis in solid tumors. Oncogene 2021; 40:5142-5152. [PMID: 34211090 PMCID: PMC8364516 DOI: 10.1038/s41388-021-01915-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Angiogenesis is essential for the sustained growth of solid tumors. Hypoxia-inducible factor 1 (HIF-1) is a master regulator of angiogenesis and constitutive activation of HIF-1 is frequently observed in human cancers. Therefore, understanding the mechanisms governing the activation of HIF-1 is critical for successful therapeutic targeting of tumor angiogenesis. Herein, we establish a new regulatory mechanism responsible for the constitutive activation of HIF-1α in cancer, irrespective of oxygen tension. PIM1 kinase directly phosphorylates HIF-1α at threonine 455, a previously uncharacterized site within its oxygen-dependent degradation domain. This phosphorylation event disrupts the ability of prolyl hydroxylases to bind and hydroxylate HIF-1α, interrupting its canonical degradation pathway and promoting constitutive transcription of HIF-1 target genes. Moreover, phosphorylation of the analogous site in HIF-2α (S435) stabilizes the protein through the same mechanism, indicating post-translational modification within the oxygen-dependent degradation domain as a mechanism of regulating the HIF-α subunits. In vitro and in vivo models demonstrate that expression of PIM1 is sufficient to stabilize HIF-1α and HIF-2α in normoxia and stimulate angiogenesis in a HIF-1-dependent manner. CRISPR mutants of HIF-1α (Thr455D) promoted increased tumor growth, proliferation, and angiogenesis. Moreover, HIF-1α-T455D xenograft tumors were refractory to the anti-angiogenic and cytotoxic effects of PIM inhibitors. These data identify a new signaling axis responsible for hypoxia-independent activation of HIF-1 and expand our understanding of the tumorigenic role of PIM1 in solid tumors.
Collapse
Affiliation(s)
- Andrea L Casillas
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | | | - Rachel K Toth
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Alva G Sainz
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Amber N Clements
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Corbin C Jensen
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Paul R Langlais
- Department of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Cindy K Miranti
- The University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Anne E Cress
- The University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Noel A Warfel
- The University of Arizona Cancer Center, Tucson, AZ, USA.
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
17
|
Cai M, Chen X, Shan J, Yang R, Guo Q, Bi X, Xu P, Shi X, Chu L, Wang L. Intermittent Hypoxic Preconditioning: A Potential New Powerful Strategy for COVID-19 Rehabilitation. Front Pharmacol 2021; 12:643619. [PMID: 33995053 PMCID: PMC8120309 DOI: 10.3389/fphar.2021.643619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a highly infectious respiratory virus, which can proliferate by invading the ACE2 receptor of host cells. Clinical studies have found that the virus can cause dyspnea, pneumonia and other cardiopulmonary system damage. In severe cases, it can lead to respiratory failure and even death. Although there are currently no effective drugs or vaccines for the prevention and treatment of COVID-19, the patient’s prognosis recovery can be effectively improved by ameliorating the dysfunction of the respiratory system, cardiovascular systems, and immune function. Intermittent hypoxic preconditioning (IHP) as a new non-drug treatment has been applied in the clinical and rehabilitative practice for treating chronic obstructive pulmonary disease (COPD), diabetes, coronary heart disease, heart failure, hypertension, and other diseases. Many clinical studies have confirmed that IHP can improve the cardiopulmonary function of patients and increase the cardiorespiratory fitness and the tolerance of tissues and organs to ischemia. This article introduces the physiological and biochemical functions of IHP and proposes the potential application plan of IHP for the rehabilitation of patients with COVID-19, so as to provide a better prognosis for patients and speed up the recovery of the disease. The aim of this narrative review is to propose possible causes and pathophysiology of COVID-19 based on the mechanisms of the oxidative stress, inflammation, and immune response, and to provide a new, safe and efficacious strategy for the better rehabilitation from COVID-19.
Collapse
Affiliation(s)
- Ming Cai
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xuan Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jieling Shan
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruoyu Yang
- College of Rehabilitation Science, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qi Guo
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xia Bi
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Ping Xu
- College of Rehabilitation Science, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiangrong Shi
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Lixi Chu
- College of Rehabilitation Science, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Shanghai Sunshine Rehabilitation Center, Shanghai, China
| | - Liyan Wang
- College of Rehabilitation Science, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
18
|
Hirota K. HIF-α Prolyl Hydroxylase Inhibitors and Their Implications for Biomedicine: A Comprehensive Review. Biomedicines 2021; 9:biomedicines9050468. [PMID: 33923349 PMCID: PMC8146675 DOI: 10.3390/biomedicines9050468] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen is essential for the maintenance of the body. Living organisms have evolved systems to secure an oxygen environment to be proper. Hypoxia-inducible factor (HIF) plays an essential role in this process; it is a transcription factor that mediates erythropoietin (EPO) induction at the transcriptional level under hypoxic environment. After successful cDNA cloning in 1995, a line of studies were conducted for elucidating the molecular mechanism of HIF activation in response to hypoxia. In 2001, cDNA cloning of dioxygenases acting on prolines and asparagine residues, which play essential roles in this process, was reported. HIF-prolyl hydroxylases (PHs) are molecules that constitute the core molecular mechanism of detecting a decrease in the partial pressure of oxygen, or hypoxia, in the cells; they can be called oxygen sensors. In this review, I discuss the process of molecular cloning of HIF and HIF-PH, which explains hypoxia-induced EPO expression; the development of HIF-PH inhibitors that artificially or exogenously activate HIF by inhibiting HIF-PH; and the significance and implications of medical intervention using HIF-PH inhibitors.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
19
|
Zhang X, Chen G, Liu Y, Sun L, Sun L, Zhao Y. Black Phosphorus-Loaded Separable Microneedles as Responsive Oxygen Delivery Carriers for Wound Healing. ACS NANO 2020; 14:5901-5908. [PMID: 32315159 DOI: 10.1021/acsnano.0c01059] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Oxygen carriers are attracting extensive interest in biomedical research and clinical applications such as wound healing, alternative blood transfusion, and acute trauma treatment. Great efforts have been devoted to the generation of oxygen carriers with special functions and properties to meet specific demands. Here, we present black phosphorus (BP)-loaded separable responsive microneedles (MNs) with oxygen carrying and controllable oxygen delivering ability for wound healing. Such MNs are composed of a polyvinyl acetate (PVA) backing layer and gelatin methacryloyl (GelMA) tips that are loaded with BP quantum dots (BP QDs) and hemoglobin (Hb). Taking advantage of the fast dissolvability of PVA, the backing layer soon disappears after the MNs are applied to skin and the noncytotoxic, biocompatible GelMA tips are left inside the skin. Due to the excellent photothermal effect of BP QDs and the reversible oxygen binding property of Hb, the local temperature of the skin will increase after near-infrared ray irradiation, resulting in the responsive oxygen release. Notably, the practical performance of such MNs has been demonstrated by treating the full-thickness cutaneous wounds of a type I diabetes rat model, indicating their potential value in wound healing and other related biomedical fields.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Guopu Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuxiao Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyu Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
20
|
Rahimi N, Azizi M, Bahari G, Narouie B, Hashemi M. Association of EGLN2 rs10680577 Polymorphism with the Risk and Clinicopathological Features of Patients with Prostate Cancer. Asian Pac J Cancer Prev 2020; 21:1221-1226. [PMID: 32458625 PMCID: PMC7541883 DOI: 10.31557/apjcp.2020.21.5.1221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Indexed: 11/25/2022] Open
Abstract
Several studies have evaluated the association between EGLN2 4-bp insertion/deletion (ins/del) polymorphism (rs10680577) and many cancers. However, up to date, no study has inspected the impact of rs10680577 polymorphism on prostate cancer (PCa) risk. This case-control study was achieved on 170 pathologically confirmed PCa patients and 196 cancer free men to inspect whether rs10680577 variant is related to the risk and clinicopathological features of patients with PCa. Genotyping was performed by mismatched polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The findings did not support an association between the variant with the risk and clinicopathological characteristics of PCa patients. When we pooled our results with six preceding studies, the findings suggested that rs10680577 variant significantly augmented the risk of overall cancer in heterozygous (OR=1.38, 95 % CI=1.26-1.52, p<0.00001, ins/del vs ins/ins), homozygous (OR=1.66, 95 % CI=1.05-2.61, p=0.029, del/del vs ins/ins), codominant (OR=1.44, 95%CI=1.32-1.58, p<0.00001, ins/del+del/del vs ins/ins), and allele (OR=1.32, 95%CI=1.18-1.49, p<0.00001, del vs ins) genetic models. Additional well designed studies with larger sample sizes are necessary to confirm our findings.
Collapse
Affiliation(s)
- Nahid Rahimi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahsa Azizi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Gholamreza Bahari
- Children and Adolescent Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Behzad Narouie
- Urology and Nephrology Research Center, Department of Urology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
21
|
Billah M, Ridiandries A, Rayner BS, Allahwala UK, Dona A, Khachigian LM, Bhindi R. Egr-1 functions as a master switch regulator of remote ischemic preconditioning-induced cardioprotection. Basic Res Cardiol 2019; 115:3. [PMID: 31823016 DOI: 10.1007/s00395-019-0763-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022]
Abstract
Despite improved treatment options myocardial infarction (MI) is still a leading cause of mortality and morbidity worldwide. Remote ischemic preconditioning (RIPC) is a mechanistic process that reduces myocardial infarction size and protects against ischemia reperfusion (I/R) injury. The zinc finger transcription factor early growth response-1 (Egr-1) is integral to the biological response to I/R, as its upregulation mediates the increased expression of inflammatory and prothrombotic processes. We aimed to determine the association and/or role of Egr-1 expression with the molecular mechanisms controlling the cardioprotective effects of RIPC. This study used H9C2 cells in vitro and a rat model of cardiac ischemia reperfusion (I/R) injury. We silenced Egr-1 with DNAzyme (ED5) in vitro and in vivo, before three cycles of RIPC consisting of alternating 5 min hypoxia and normoxia in cells or hind-limb ligation and release in the rat, followed by hypoxic challenge in vitro and I/R injury in vivo. Post-procedure, ED5 administration led to a significant increase in infarct size compared to controls (65.90 ± 2.38% vs. 41.00 ± 2.83%, p < 0.0001) following administration prior to RIPC in vivo, concurrent with decreased plasma IL-6 levels (118.30 ± 4.30 pg/ml vs. 130.50 ± 1.29 pg/ml, p < 0.05), downregulation of the cardioprotective JAK-STAT pathway, and elevated myocardial endothelial dysfunction. In vitro, ED5 administration abrogated IL-6 mRNA expression in H9C2 cells subjected to RIPC (0.95 ± 0.20 vs. 6.08 ± 1.40-fold relative to the control group, p < 0.05), resulting in increase in apoptosis (4.76 ± 0.70% vs. 2.23 ± 0.34%, p < 0.05) and loss of mitochondrial membrane potential (0.57 ± 0.11% vs. 1.0 ± 0.14%-fold relative to control, p < 0.05) in recipient cells receiving preconditioned media from the DNAzyme treated donor cells. This study suggests that Egr-1 functions as a master regulator of remote preconditioning inducing a protective effect against myocardial I/R injury through IL-6-dependent JAK-STAT signaling.
Collapse
Affiliation(s)
- M Billah
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia.
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Life Sciences, Independent University Bangladesh, Dhaka, Bangladesh.
| | - A Ridiandries
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - B S Rayner
- Inflammation Group, Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| | - U K Allahwala
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - A Dona
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - L M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - R Bhindi
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, Level 12, Royal North Shore Hospital, Cnr Reserve Rd and Westbourne, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
22
|
Peng Q, Li H, Chen J, Wang Y, Tang Z. Oral submucous fibrosis in Asian countries. J Oral Pathol Med 2019; 49:294-304. [PMID: 31310693 DOI: 10.1111/jop.12924] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/19/2022]
Abstract
Oral submucous fibrosis (OSF) is a chronic, insidious, and progressive oral mucosal disease that affects entire oral cavity and sometimes pharynx. This oral potentially malignant disorder has a high rate of malignant transformation (7%-30%) to oral squamous cell carcinoma (OSCC), posing global problems for public health. Due to enormous efforts dedicated to this disease in the past decades, there have been significant advances in identification of its etiology and pathogenesis as well as development of corresponding therapeutic approaches, in spite of several challenges. This study reviewed the existing literature concerning OSF in Asian countries, encompassing its etiology, histopathology, pathogenesis, clinical manifestations, diagnosis and differential diagnosis, and treatments. For improving treatment of OSF, the multifactorial etiology analysis, incorporation of effective molecular pathways, cytokines and cells for mechanism illustration, and integration of multidisciplinary modalities were also expounded to guide future research and clinical practice.
Collapse
Affiliation(s)
- Qian Peng
- Xiangya Stomatological Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hongyuan Li
- Xiangya Stomatological Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| | - Juan Chen
- Xiangya Stomatological Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yuehong Wang
- Xiangya Stomatological Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Xiangya Stomatological Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
23
|
Lee Chong T, Ahearn EL, Cimmino L. Reprogramming the Epigenome With Vitamin C. Front Cell Dev Biol 2019; 7:128. [PMID: 31380368 PMCID: PMC6646595 DOI: 10.3389/fcell.2019.00128] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/02/2019] [Indexed: 12/19/2022] Open
Abstract
The erasure of epigenetic modifications across the genome of somatic cells is an essential requirement during their reprogramming into induced pluripotent stem cells (iPSCs). Vitamin C plays a pivotal role in remodeling the epigenome by enhancing the activity of Jumonji-C domain-containing histone demethylases (JHDMs) and the ten-eleven translocation (TET) proteins. By maintaining differentiation plasticity in culture, vitamin C also improves the quality of tissue specific stem cells derived from iPSCs that are highly sought after for use in regenerative medicine. The ability of vitamin C to potentiate the activity of histone and DNA demethylating enzymes also has clinical application in the treatment of cancer. Vitamin C deficiency has been widely reported in cancer patients and has recently been shown to accelerate cancer progression in disease models. Therapies involving high-dose vitamin C administration are currently gaining traction in the treatment of epigenetic dysregulation, by targeting aberrant histone and DNA methylation patterns associated with cancer progression.
Collapse
Affiliation(s)
- Taylor Lee Chong
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Emily L Ahearn
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Luisa Cimmino
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
24
|
Iglesias P, Penas C, Barral-Cagiao L, Pazos E, Costoya JA. A Bio-inspired Hypoxia Sensor using HIF1a-Oxygen-Dependent Degradation Domain. Sci Rep 2019; 9:7117. [PMID: 31068630 PMCID: PMC6506541 DOI: 10.1038/s41598-019-43618-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/24/2019] [Indexed: 12/31/2022] Open
Abstract
Functional imaging has become an important tool in oncology because it not only provides information about the size and localization of the tumour, but also about the pathophysiological features of the tumoural cells. One of the characteristic features of some tumour types is that their fast growth leads to deficient intratumoral vascularization, which results in low oxygen availability. To overcome this lack of oxygen, tumoural cells activate the neoangiogenic program by upregulating the transcription factor HIF-1α. Herein we report a non-invasive in vitro detection method of hypoxia using designed fluorescent peptide probes based on the oxygen-dependent degradation domain of HIF-1α. The fluorescent probe retains the oxygen-sensing capability of HIF-1α, so that it is stabilized under hypoxia and readily degraded by the proteasome under normoxia, thus providing direct information of the cellular oxygen availability.
Collapse
Affiliation(s)
- Pablo Iglesias
- Molecular Oncology Laboratory. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristina Penas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Laura Barral-Cagiao
- Molecular Oncology Laboratory. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Elena Pazos
- Departamento de Química, Facultade de Ciencias and Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, 15071, A Coruña, Spain.
| | - Jose A Costoya
- Molecular Oncology Laboratory. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
25
|
Hirota K. An intimate crosstalk between iron homeostasis and oxygen metabolism regulated by the hypoxia-inducible factors (HIFs). Free Radic Biol Med 2019; 133:118-129. [PMID: 30053508 DOI: 10.1016/j.freeradbiomed.2018.07.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/24/2022]
Abstract
Oxygen and iron are among the most abundant elements and have significant roles in human biology. Iron is essential for oxygen transport and is a component of molecular O2-carrying proteins, such as hemoglobin and myoglobin. Iron is also a constituent of redox enzymes and can occupy multiple oxidation states. An elaborate system has evolved to stringently regulate the concentrations of both, free iron and oxygen, in various sites of the body. The final destination for iron and oxygen in the cells is the mitochondria. The mitochondria require substantial amounts of iron for heme synthesis and maturation of iron-sulfur clusters, and oxygen, as the electron acceptor in oxidative phosphorylation. Therefore, the balance between the control of iron availability and the physiology of hypoxic responses is critical for maintaining cell homeostasis. Several lines of study have clearly demonstrated that the transcription factors, hypoxia-inducible factors (HIFs), play a central role in cellular adaptation to critically low oxygen levels in both normal and compromised tissues. It has also been shown that several target genes of HIFs are involved in iron homeostasis, reflecting the molecular links between oxygen homeostasis and iron metabolism. Furthermore, HIF activation is modulated by intracellular iron, through regulation of hydroxylase activity, which requires iron as a cofactor. In addition, HIF-2α translation is controlled by iron regulatory protein (IRP) activity, providing another level of interdependence between iron and oxygen homeostasis.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan.
| |
Collapse
|
26
|
Hashemi M, Tabasi F, Ansari H. 4-bp insertion/deletion polymorphism within the promoter of EGLN2 gene is associated with susceptibility to cancer in Asian population: Evidence from a meta-analysis. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
27
|
Maugeri G, D'Amico AG, Rasà DM, Saccone S, Federico C, Cavallaro S, D'Agata V. PACAP and VIP regulate hypoxia-inducible factors in neuroblastoma cells exposed to hypoxia. Neuropeptides 2018; 69:84-91. [PMID: 29699729 DOI: 10.1016/j.npep.2018.04.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/13/2018] [Accepted: 04/15/2018] [Indexed: 12/20/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two related peptides acting as neurotransmitters/neuromodulators in central and peripheral nervous system. They are also involved in cancer showing a controversial role. Particulary, they are implicated in neuroblastoma differentiation (NB). This pediatric tumor can evolve to a malignant metastatic disease or spontaneously regress towards a benign form, known as ganglioneuroblastoma/ganglioneuroma. A negative hallmark of neoplasia progression is represented by hypoxic microenvironment. Low oxygen tension induces activation of hypoxia-inducible factors (HIFs) promoting cells proliferation and metastasis formation. Moreover, HIFs trigger vascular endothelial growth factor (VEGF) release favouring high-risk NB phenotype development. In the present work, we have investigated for the first time, if PACAP and VIP interfere with NB differentiation through modulation of hypoxic/angiogenic process. To this end, we analyzed their effect in malignant undifferentiated and all-trans retinoic acid (RA) differentiated SH-SY5Y cells, representing the benign form of this tumor. Our results have suggested tha both peptides, but predominantly VIP, induce NB differentiation into benign form by regulating HIFs, VEGF and VEGFRs expression and distribution. All these data give new insight regarding PACAP/VIP regulatory role in NB progression.
Collapse
Affiliation(s)
- Grazia Maugeri
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agata Grazia D'Amico
- Department of Human Science and Promotion of quality of Life, San Raffaele Open University of Rome, Italy
| | - Daniela Maria Rasà
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Saccone
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | | | - Velia D'Agata
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
28
|
Wang CC, Ying L, Barnes EA, Adams ES, Kim FY, Engel KW, Alvira CM, Cornfield DN. Pulmonary artery smooth muscle cell HIF-1α regulates endothelin expression via microRNA-543. Am J Physiol Lung Cell Mol Physiol 2018; 315:L422-L431. [PMID: 29745253 DOI: 10.1152/ajplung.00475.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary artery smooth muscle cells (PASMCs) express endothelin (ET-1), which modulates the pulmonary vascular response to hypoxia. Although cross-talk between hypoxia-inducible factor-1α (HIF-1α), an O2-sensitive transcription factor, and ET-1 is established, the cell-specific relationship between HIF-1α and ET-1 expression remains incompletely understood. We tested the hypotheses that in PASMCs 1) HIF-1α expression constrains ET-1 expression, and 2) a specific microRNA (miRNA) links HIF-1α and ET-1 expression. In human (h)PASMCs, depletion of HIF-1α with siRNA increased ET-1 expression at both the mRNA and protein levels ( P < 0.01). In HIF-1α-/- murine PASMCs, ET-1 gene and protein expression was increased ( P < 0.0001) compared with HIF-1α+/+ cells. miRNA profiles were screened in hPASMCs transfected with siRNA-HIF-1α, and RNA hybridization was performed on the Agilent (Santa Clara, CA) human miRNA microarray. With HIF-1α depletion, miRNA-543 increased 2.4-fold ( P < 0.01). In hPASMCs, miRNA-543 overexpression increased ET-1 gene ( P < 0.01) and protein ( P < 0.01) expression, decreased TWIST gene expression ( P < 0.05), and increased ET-1 gene and protein expression, compared with nontargeting controls ( P < 0.01). Moreover, we evaluated low passage hPASMCs from control and patients with idiopathic pulmonary arterial hypertension (IPAH). Compared with controls, protein expression of HIF-1α and Twist-related protein-1 (TWIST1) was decreased ( P < 0.05), and miRNA-543 and ET-1 expression increased ( P < 0.001) in hPASMCs from patients with IPAH. Thus, in PASMCs, loss of HIF-1α increases miRNA-543, which decreases Twist expression, leading to an increase in PASMC ET-1 expression. This previously undescribed link between HIF-1α and ET-1 via miRNA-543 mediated Twist suppression represents another layer of molecular regulation that might determine pulmonary vascular tone.
Collapse
Affiliation(s)
- Ching-Chia Wang
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Department of Pediatrics, National Taiwan University Children Hospital, National Taiwan University Medical College , Taipei , Taiwan
| | - Lihua Ying
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University Medical School , Stanford, California
| | - Elizabeth A Barnes
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University Medical School , Stanford, California
| | - Eloa S Adams
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Kaiser Oakland, Oakland, California
| | - Francis Y Kim
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Milwaukee Children's Hospital, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Karl W Engel
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California
| | - Cristina M Alvira
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Division of Critical Care Medicine, Department of Pediatrics, Stanford University Medical School , Stanford, California
| | - David N Cornfield
- Center for Excellence in Pulmonary Biology, Stanford University Medical School , Stanford, California.,Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University Medical School , Stanford, California.,Division of Critical Care Medicine, Department of Pediatrics, Stanford University Medical School , Stanford, California
| |
Collapse
|
29
|
Serocki M, Bartoszewska S, Janaszak-Jasiecka A, Ochocka RJ, Collawn JF, Bartoszewski R. miRNAs regulate the HIF switch during hypoxia: a novel therapeutic target. Angiogenesis 2018; 21:183-202. [PMID: 29383635 PMCID: PMC5878208 DOI: 10.1007/s10456-018-9600-2] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
The decline of oxygen tension in the tissues below the physiological demand leads to the hypoxic adaptive response. This physiological consequence enables cells to recover from this cellular insult. Understanding the cellular pathways that mediate recovery from hypoxia is therefore critical for developing novel therapeutic approaches for cardiovascular diseases and cancer. The master regulators of oxygen homeostasis that control angiogenesis during hypoxia are hypoxia-inducible factors (HIFs). HIF-1 and HIF-2 function as transcriptional regulators and have both unique and overlapping target genes, whereas the role of HIF-3 is less clear. HIF-1 governs the acute adaptation to hypoxia, whereas HIF-2 and HIF-3 expressions begin during chronic hypoxia in human endothelium. When HIF-1 levels decline, HIF-2 and HIF-3 increase. This switch from HIF-1 to HIF-2 and HIF-3 signaling is required in order to adapt the endothelium to prolonged hypoxia. During prolonged hypoxia, the HIF-1 levels and activity are reduced, despite the lack of oxygen-dependent protein degradation. Although numerous protein factors have been proposed to modulate the HIF pathways, their application for HIF-targeted therapy is rather limited. Recently, the miRNAs that endogenously regulate gene expression via the RNA interference (RNAi) pathway have been shown to play critical roles in the hypoxia response pathways. Furthermore, these classes of RNAs provide therapeutic possibilities to selectively target HIFs and thus modulate the HIF switch. Here, we review the significance of the microRNAs on the relationship between the HIFs under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Marcin Serocki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdańsk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdańsk, Poland
| | - Renata J Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdańsk, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rafał Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdańsk, Poland.
| |
Collapse
|
30
|
Hashemi M, Danesh H, Bizhani F, Sattarifard H, Hashemi SM, Bahari G. Detection of a 4-bp Insertion/deletion Polymorphism within the Promoter of EGLN2 Using Mismatch PCR-RFLP and Its Association with Susceptibility to Breast Cancer. Asian Pac J Cancer Prev 2018; 19:923-926. [PMID: 29693343 PMCID: PMC6031807 DOI: 10.22034/apjcp.2018.19.4.923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has been shown that a 4-bp insertion/deletion (ins/del) polymorphism of EGLN2 influences the risk of several cancers. However, to date, no study has inspected the impact of the 4-bp ins/del polymorphism on breast cancer (BC) risk. A case-control study, including 134 breast cancer patients and 154 healthy women, was here conducted to examine the possible association between EGLN2 4-bp ins/del polymorphism and BC risk in a southeast Iranian population. A mismatched polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was designed for genotyping of the variant. Our findings did not support any association between the 4-bp ins/del polymorphism and the risk of BC in the codominant, dominant, recessive and allele inheritance models tested. When links between the EGLN2 4-bp ins/del polymorphism and clinicopathological characteristics of the patients were evaluate the variant was only associated with HER2 status. More studies with larger sample sizes and diverse ethnicities are warranted to verify our finding.
Collapse
Affiliation(s)
- Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | | | | | | | | | |
Collapse
|
31
|
Blewett TA, Simon RA, Turko AJ, Wright PA. Copper alters hypoxia sensitivity and the behavioural emersion response in the amphibious fish Kryptolebias marmoratus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 189:25-30. [PMID: 28575749 DOI: 10.1016/j.aquatox.2017.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/03/2017] [Accepted: 05/16/2017] [Indexed: 06/07/2023]
Abstract
Elevated levels of metals have been reported in mangrove ecosystems worldwide. Mangrove fishes also routinely experience severe environmental stressors, such as hypoxia. In the amphibious fish Kryptolebias marmoratus (mangrove rivulus), a key behavioural response to avoid aquatic stress is to leave water (emersion). We hypothesized that copper (Cu) exposure would increase the sensitivity of this behavioural hypoxia avoidance response due to histopathological effects of Cu on gill structure and function. K. marmoratus were exposed to either control (no added Cu) or Cu (300μg/L) for 96h. Following this period, fish were exposed to an acute hypoxic challenge (decline in dissolved oxygen to ∼0% over 15min), and the emersion response was recorded. Gills were examined for histological changes. Fish exposed to Cu emersed at a higher dissolved oxygen level (7.5±0.6%), relative to the control treatment group (5.8±0.4%). Histological analysis showed that the gill surface area increased and the interlamellar cell mass (ILCM) was reduced following Cu exposure, contrary to our prediction. Overall, these data indicate that Cu induces hypoxia-like changes to gill morphology and increases the sensitivity of the hypoxia emersion response.
Collapse
Affiliation(s)
- Tamzin A Blewett
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario, N2L 3C5, Canada.
| | - Robyn A Simon
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Andy J Turko
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Patricia A Wright
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| |
Collapse
|
32
|
Wu K, Hu M, Chen Z, Xiang F, Chen G, Yan W, Peng Q, Chen X. Asiatic acid enhances survival of human AC16 cardiomyocytes under hypoxia by upregulating miR-1290. IUBMB Life 2017; 69:660-667. [PMID: 28686797 DOI: 10.1002/iub.1648] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/06/2017] [Indexed: 02/05/2023]
Abstract
Asiatic acid (AA) could attenuate ischemia/reperfusion induced myocardial apoptosis through upregulating the Akt/GSK-3β/HIF-1α pathway. HIF-3α is a negative regulator of HIF-1α, whose mRNA is a potential target of miR-1290. AA could upregulate miR-1290 in non-small-cell lung cancer A549 cells. This work aimed to investigate whether AA could inhibit hypoxia induced cardiomyocyte apoptosis through regulating the miR-1290/HIF3A/HIF-1α axis. The AC16 human myocardial cell line cultured under normoxic or hypoxic conditions was treated with various doses of AA for 24 h. Afterwards cell viability, apoptosis and the expression of miR-1290, HIF3A, and HIF1A were evaluated. Cells transfected with miR-1290 mimic or inhibitor were used to determine the role of miR-1290 in the anti-apoptosis effect of AA and the expression of HIF3A and HIF1A. Dual luciferase assay was performed to confirm miR-1290 targeting of HIF3A. HIF3A overexpression was achieved by transfection of HIF3A1 overexpressing lentivirus, and its effect on miR-1290 and AA-regulated survival of cardiomyocytes was evaluated. AA treatment protected cardiomyocytes from hypoxia-induced apoptosis and upregulated miR-1290 and HIF1A, but downregulated HIF3A under hypoxia. The protective effect of AA was abolished by miR-1290 knockdown, whereas enhanced by miR-1290 overexpression. In addition, miR-1290 knockdown increased HIF1A expression, but reduced HIF3A expression in cardiomyocytes. Dual luciferase assay confirmed miR-1290 direct targeting the 3' UTR of HIF3A. HIF3A overexpression counteracted the anti-apoptosis effect of AA or miR-1290. In conclusion, AA can protect cardiomyocytes against hypoxia-induced apoptosis through regulating the miR-1290/HIF3A/HIF-1α axis, and miR-1290 may be a potential target in the prevention of myocardial ischemia-reperfusion injury. © 2017 IUBMB Life, 69(9):660-667, 2017.
Collapse
Affiliation(s)
- Kai Wu
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Min Hu
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Zejin Chen
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Feixiang Xiang
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Guojian Chen
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Weihong Yan
- Department of Health Management, 903 Hospital, Mianyang, Sichuan, China
| | - Qing Peng
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoping Chen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Zeng Y, Tao L, Ma J, Han L, Lv Y, Hui P, Zhang H, Ma K, Xiao B, Shi Q, Xu H, Chen L. DUSP1 and KCNJ2 mRNA upregulation can serve as a biomarker of mechanical asphyxia-induced death in cardiac tissue. Int J Legal Med 2017. [PMID: 28624985 DOI: 10.1007/s00414-017-1616-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The incidence of death by asphyxia is second to the incidence of death by mechanical injury; however, death by mechanical asphyxia may be difficult to prove in court, particularly in cases in which corpses do not exhibit obvious signs of asphyxia. To identify a credible biomarker of asphyxia, we first examined the expression levels of 47,000 mRNAs in human cardiac tissue specimens from individuals who died of mechanical asphyxia and compared the expression levels with the levels of the corresponding mRNAs in specimens from individuals who died of craniocerebral injury using microarray. We selected 119 differentially expressed mRNAs, examined the expression levels of these mRNAs in 44 human cardiac tissue specimens of individuals who died of mechanical asphyxia, craniocerebral injury, hemorrhagic shock, or other causes. That the expression of dual-specificity phosphatase 1 (DUSP1) and potassium voltage-gated channel subfamily J member 2 (KCNJ2) was upregulated in human cardiac tissues from the mechanical asphyxia group compared with control tissues, regardless of age, environmental temperature, and postmortem interval (PMI), indicating that DUSP1 and KCNJ2 may be associated with mechanical asphyxia-induced death and can thus serve as useful biomarkers of death by mechanical asphyxia.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Li Tao
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Jianlong Ma
- Criminal Investigation Department of Shenzhen Public Security Bureau, Shenzhen Insitute of Criminal Science and Technology, Shenzhen, 518000, China
| | - Liujun Han
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Yehui Lv
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China.,Shanghai University of Medicine & Health Sciences, 279 ZhouzhuHwy, Shanghai, 201318, People's Republic of China
| | - Pan Hui
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Heng Zhang
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Kaijun Ma
- Forensic laboratory, Criminal Science and Technology Institute, Shanghai Public Security Bureau, 803 North Zhongshan Road, Shanghai, 200082, People's Republic of China
| | - Bi Xiao
- Forensic laboratory, Criminal Science and Technology Institute, Shanghai Public Security Bureau, 803 North Zhongshan Road, Shanghai, 200082, People's Republic of China
| | - Qun Shi
- Forensic laboratory, Criminal Science and Technology Institute, Shanghai Public Security Bureau, 803 North Zhongshan Road, Shanghai, 200082, People's Republic of China
| | - Hongmei Xu
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China.
| | - Long Chen
- Department of Forensic Medicine, Schoolof Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
34
|
Jankovic A, Korac A, Buzadzic B, Stancic A, Otasevic V, Ferdinandy P, Daiber A, Korac B. Targeting the NO/superoxide ratio in adipose tissue: relevance to obesity and diabetes management. Br J Pharmacol 2017; 174:1570-1590. [PMID: 27079449 PMCID: PMC5446578 DOI: 10.1111/bph.13498] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin sensitivity and metabolic homeostasis depend on the capacity of adipose tissue to take up and utilize excess glucose and fatty acids. The key aspects that determine the fuel-buffering capacity of adipose tissue depend on the physiological levels of the small redox molecule, nitric oxide (NO). In addition to impairment of NO synthesis, excessive formation of the superoxide anion (О2•- ) in adipose tissue may be an important interfering factor diverting the signalling of NO and other reactive oxygen and nitrogen species in obesity, resulting in metabolic dysfunction of adipose tissue over time. Besides its role in relief from superoxide burst, enhanced NO signalling may be responsible for the therapeutic benefits of different superoxide dismutase mimetics, in obesity and experimental diabetes models. This review summarizes the role of NO in adipose tissue and highlights the effects of NO/О2•- ratio 'teetering' as a promising pharmacological target in the metabolic syndrome. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron MicroscopyUniversity of BelgradeBelgradeSerbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Péter Ferdinandy
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
| | - Andreas Daiber
- Center for Cardiology ‐ Cardiology 1, Molecular CardiologyUniversity Medical CenterMainzGermany
| | - Bato Korac
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| |
Collapse
|
35
|
Maugeri G, D'Amico AG, Reitano R, Magro G, Cavallaro S, Salomone S, D'Agata V. PACAP and VIP Inhibit the Invasiveness of Glioblastoma Cells Exposed to Hypoxia through the Regulation of HIFs and EGFR Expression. Front Pharmacol 2016; 7:139. [PMID: 27303300 PMCID: PMC4885839 DOI: 10.3389/fphar.2016.00139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/12/2016] [Indexed: 11/30/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal
peptide (VIP) through the binding of vasoactive intestinal peptide receptors (VIPRs),
perform a wide variety of effects in human cancers, including glioblastoma multiforme
(GBM). This tumor is characterized by extensive areas of hypoxia, which triggers the
expression of hypoxia-inducible factors (HIFs). HIFs not only mediate angiogenesis
but also tumor cell migration and invasion. Furthermore, HIFs activation is linked to
epidermal growth factor receptor (EGFR) overexpression. Previous studies have shown
that VIP interferes with the invasive nature of gliomas by regulating cell migration.
However, the role of VIP family members in GBM infiltration under low oxygen tension
has not been clarified yet. Therefore, in the present study we have investigated, for
the first time, the molecular mechanisms involved in the anti-invasive effect of
PACAP or VIP in U87MG glioblastoma cells exposed to hypoxia induced by treatment with
desferrioxamine (DFX). The results suggest that either PACAP or VIP exert an
anti-infiltrative effect under low oxygen tension by modulating HIFs and EGFR
expression, key elements involved in cell migration and angiogenesis. These peptides
act through the inhibition of PI3K/Akt and MAPK/ERK signaling pathways, which are
known to have a crucial role in HIFs regulation.
Collapse
Affiliation(s)
- Grazia Maugeri
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Agata Grazia D'Amico
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy; San Raffaele Open University of RomeRome, Italy
| | - Rita Reitano
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Gaetano Magro
- Section of Anatomic Pathology, Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", University of Catania Catania, Italy
| | | | - Salvatore Salomone
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Velia D'Agata
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| |
Collapse
|
36
|
Lee YH, Lee JM, Kim SG, Lee YS. Synthesis and biological evaluation of 1,2-dithiol-3-thiones and pyrrolo[1,2-a]pyrazines as novel hypoxia inducible factor-1 (HIF-1) inhibitor. Bioorg Med Chem 2016; 24:2843-51. [PMID: 27157007 DOI: 10.1016/j.bmc.2016.04.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/24/2016] [Accepted: 04/25/2016] [Indexed: 11/25/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key transcription factor which is strongly associated with tumor survival, progression, and therapeutic resistance. Accordingly, it has been suggested that the inhibition of the HIF-1 pathway can suppress tumor, and it has become an important therapeutic target. In present study, oltipraz, its metabolite M2, and their derivatives were synthesized and evaluated as HIF-1α inhibitors. Among the synthesized, benzyl-substituted pyrrolo[1,2-a]pyrazine 2g most potently inhibited HIF-1α protein accumulation (81% at 10μM) and VEGF, GLUT-1 transcription (77% and 92% at 10μM, respectively).
Collapse
Affiliation(s)
- Young Hun Lee
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jung Min Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Geon Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong Sup Lee
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea; Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
37
|
Molecular response and association analysis of Megalobrama amblycephala fih-1 with hypoxia. Mol Genet Genomics 2016; 291:1615-24. [PMID: 27112926 DOI: 10.1007/s00438-016-1208-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 04/13/2016] [Indexed: 12/19/2022]
Abstract
Hypoxia is one of the most important environmental factors which affect fish growth, development and survival, but regulation mechanisms of hypoxia in fish remain unclear. Therefore, to further understand molecular functions of factor inhibiting HIF-1 (Fih-1), an essential hypoxia sensor, the full-length cDNA of fih-1 was cloned from Megalobrama amblycephala, a hypoxia-sensitive cyprinid fish. The deduced amino acid sequence showed high homology with that of other vertebrates, and all structural and functional domains were highly conserved. The mRNA level in different tissues and developmental stages indicated that M. amblycephala fih-1 expression was higher in liver and muscle, followed by gill, intestine and spleen. During embryogenesis, the fih-1 mRNA was highly expressed in the early embryonic development, then decreased to a very low level, and maintained a relative high level of expression after hatching. In most tissues, the fih-1 mRNA was down-regulated at 2 h but up-regulated at 4 h after hypoxia treatment. In addition, the promoter sequence of M. amblycephala fih-1 was obtained using thermal asymmetric interlaced PCR. Three single nucleotide polymorphism (SNP) sites were found in the cDNA and promoter sequences, and identified significant association with hypoxia trait by correlation analysis in hypoxia-sensitive group and hypoxia-tolerant group. These results demonstrated that M. amblycephala fih-1 plays important roles in embryo development and hypoxia response, which will contribute to systematic understanding of the molecular mechanisms of fish in response to hypoxia, and provide help for fish genetic breeding with hypoxia-tolerant strains or breeds.
Collapse
|
38
|
Wang Y, Lu H, Chen Y, Luo Y. The association of angiotensin-converting enzyme gene insertion/deletion polymorphisms with adaptation to high altitude: A meta-analysis. J Renin Angiotensin Aldosterone Syst 2016; 17:1470320315627410. [PMID: 27009284 PMCID: PMC5843938 DOI: 10.1177/1470320315627410] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/22/2015] [Indexed: 12/20/2022] Open
Abstract
Background: Fluid retention is linked to the physiology and pathophysiology of humans at high altitude (HA). The angiotensin-converting enzyme (ACE) gene plays a role in the regulation of plasma volume and vascular tone. Materials and methods: In this meta-analysis, eligible studies published before 1 September 2015 that focused on the association between the ACE insertion/deletion (I/D) polymorphism and HA adaption were identified by searching the PubMed, Web of Science, Embase and Medline online databases. We used a fixed-effects model and assessed the study qualities multiple times. Results: The seven selected studies included a total of 582 HA-native individuals and 497 low-altitude controls, and these subjects were analyzed for the ACE I/D gene polymorphism. A significant association was found between the ACE DD genotype and HA maladaptation. The results for genotype DD versus ID + II were as follows: Odds ratio (OR) = 0.46; 95% CI 0.31–0.70; p = 0.0002. The results for genotype ID versus DD were as follows: OR = 1.97; 95% CI 1.27–3.06; p = 0.002. Conclusions: Our findings suggested that the DD genotype of ACE is a risk factor for HA maladaptation and that the presence of fewer ACE DD allele carriers in a population indicates a greater ability of that population to adapt to HA.
Collapse
Affiliation(s)
- Yuxiao Wang
- Department of Military Medical Geography, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China Battalion 5 Cadet Brigade, Third Military Medical University, Chongqing, China Key Laboratory of High-altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Hongxiang Lu
- Department of Military Medical Geography, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China Battalion 5 Cadet Brigade, Third Military Medical University, Chongqing, China Key Laboratory of High-altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Yu Chen
- Key Laboratory of High-altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Yongjun Luo
- Department of Military Medical Geography, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China Key Laboratory of High-altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China
| |
Collapse
|
39
|
miR-429 regulates the transition between Hypoxia-Inducible Factor (HIF)1A and HIF3A expression in human endothelial cells. Sci Rep 2016; 6:22775. [PMID: 26954587 PMCID: PMC4782134 DOI: 10.1038/srep22775] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/23/2016] [Indexed: 12/22/2022] Open
Abstract
Hypoxia-inducible factors (HIF) are heterodimeric transcription factors that allow cells to adapt and survive during hypoxia. Regulation of HIF1A and HIF2A mRNA is well characterized, whereas HIF3A mRNA regulation and function are less clear. Using RNA-Seq analysis of primary human umbilical vein endothelial cells, we found two isoforms of HIF3A were expressed, HIF3A2 and HIF3A3. Comparing HIF3A expression profiles to HIF1A mRNA during 48 hours of hypoxia revealed that HIF1A message peaked at 4 hours, whereas HIF3A expression increased while HIF1A was decreasing. Given that HIF1A mRNA is regulated by miR-429, we tested miR-429 effects on both HIF3A isoforms and found that they too were regulated by miR-429. Analysis of a HIF-3 target, DNA-damage-inducible transcript 4, a key survival gene, indicated that DDIT4 mRNA is induced by HIF-3 and negatively regulated by miR-429 through miR-429’s actions on HIF3A message. This provides a compelling model for how hypoxia-induced miR-429 regulates the switch between HIF-1 adaptive responses to HIF-3 survival responses by rapidly decreasing HIF1A levels while simultaneously slowing the progression of HIF3A expression until the miR-429 levels drop below normoxic levels. Since HIF-1 drives HIF3A and miR-429 expression, this establishes a regulatory network in which miR-429 plays a pivotal role.
Collapse
|
40
|
Maugeri G, D'Amico AG, Reitano R, Saccone S, Federico C, Cavallaro S, D'Agata V. Parkin modulates expression of HIF-1α and HIF-3α during hypoxia in gliobastoma-derived cell lines in vitro. Cell Tissue Res 2016; 364:465-474. [PMID: 26742768 DOI: 10.1007/s00441-015-2340-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/28/2015] [Accepted: 12/02/2015] [Indexed: 12/11/2022]
Abstract
Mutation of the Parkin gene causes an autosomal recessive juvenile-onset form of Parkinson's disease. However, recently, it has been also linked to a wide variety of malignancies, including glioblastoma multiforme (GBM). In this pathology, Parkin exhibits a tumor suppressor role by mitigating the proliferation rate in both in vitro and in vivo models. However, Parkin involvement in the hypoxic process has not as yet been investigated. GBM is the most common and aggressive primary brain tumor in adults and is characterized by hypoxic areas. The low oxygen supply causes the expression of hypoxia-inducible factors (HIFs) leading to an accumulation of pro-angiogenic factors and tumoral invasiveness. We assess the relationship between Parkin and two HIFs expressed during hypoxic conditions, namely HIF-1α and HIF-3α. Our data show that Parkin is downregulated under hypoxia and that it interferes with HIF expression based on cellular oxygen tension. These results suggest a role for the involvement of Parkin in GBM, although further studies will be needed to understand the mechanism by which it modulates HIF-1α and HIF-3α expression.
Collapse
Affiliation(s)
- Grazia Maugeri
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S.Sofia 87, 95123, Catania, Italy
| | - Agata Grazia D'Amico
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S.Sofia 87, 95123, Catania, Italy
- San Raffaele Telematic University of Rome, Rome, Italy
| | - Rita Reitano
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S.Sofia 87, 95123, Catania, Italy
| | - Salvatore Saccone
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Sebastiano Cavallaro
- Functional Genomics Unit, Institute of Neurological Sciences, Italian National Research Council, Catania, Italy
| | - Velia D'Agata
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S.Sofia 87, 95123, Catania, Italy.
| |
Collapse
|
41
|
Nüsken E, Herrmann Y, Wohlfarth M, Goecke T, Appel S, Schneider H, Dötsch J, Nüsken K. Strong hypoxia reduces leptin synthesis in purified primary human trophoblasts. Placenta 2015; 36:427-32. [DOI: 10.1016/j.placenta.2015.01.191] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/20/2015] [Accepted: 01/24/2015] [Indexed: 12/20/2022]
|
42
|
Ehsan SM, George SC. Vessel network formation in response to intermittent hypoxia is frequency dependent. J Biosci Bioeng 2015; 120:347-50. [PMID: 25735591 DOI: 10.1016/j.jbiosc.2015.01.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/09/2014] [Accepted: 01/15/2015] [Indexed: 10/23/2022]
Abstract
A combined experimental and mathematical model of intermittent hypoxia (IH) conditioned engineered tissue was used to characterize the effects of IH on the formation of in vitro vascular networks. Results showed that the frequency of hypoxic oscillations has pronounced influence on the vascular response of endothelial cells and fibroblasts.
Collapse
Affiliation(s)
- Seema M Ehsan
- Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, 2400 Engineering Hall, Irvine, CA 92697, USA
| | - Steven C George
- Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, 2400 Engineering Hall, Irvine, CA 92697, USA; Biomedical Engineering, University of California, 3120 Natural Sciences II, Irvine, CA 92697, USA; Department of Medicine, University of California, 333 City Blvd. West, Suite 400, Orange, CA 92868, USA.
| |
Collapse
|
43
|
Jeon YJ, Kim HS, Song KS, Han HJ, Park SH, Chang W, Lee MY. Protective effect of dieckol against chemical hypoxia-induced cytotoxicity in primary cultured mouse hepatocytes. Drug Chem Toxicol 2014; 38:180-7. [DOI: 10.3109/01480545.2014.928719] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
El-Hattab AW, Emrick LT, Chanprasert S, Craigen WJ, Scaglia F. Mitochondria: Role of citrulline and arginine supplementation in MELAS syndrome. Int J Biochem Cell Biol 2014; 48:85-91. [DOI: 10.1016/j.biocel.2013.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 11/14/2013] [Accepted: 12/26/2013] [Indexed: 12/18/2022]
|
45
|
Kubota S, Fukumoto Y, Ishibashi K, Soeda S, Kubota S, Yuki R, Nakayama Y, Aoyama K, Yamaguchi N, Yamaguchi N. Activation of the prereplication complex is blocked by mimosine through reactive oxygen species-activated ataxia telangiectasia mutated (ATM) protein without DNA damage. J Biol Chem 2014; 289:5730-46. [PMID: 24421316 DOI: 10.1074/jbc.m113.546655] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mimosine is an effective cell synchronization reagent used for arresting cells in late G1 phase. However, the mechanism underlying mimosine-induced G1 cell cycle arrest remains unclear. Using highly synchronous cell populations, we show here that mimosine blocks S phase entry through ATM activation. HeLa S3 cells are exposed to thymidine for 15 h, released for 9 h by washing out the thymidine, and subsequently treated with 1 mM mimosine for a further 15 h (thymidine → mimosine). In contrast to thymidine-induced S phase arrest, mimosine treatment synchronizes >90% of cells at the G1-S phase boundary by inhibiting the transition of the prereplication complex to the preinitiation complex. Mimosine treatment activates ataxia telangiectasia mutated (ATM)/ataxia telangiectasia and Rad3-related (ATR)-mediated checkpoint signaling without inducing DNA damage. Inhibition of ATM activity is found to induce mimosine-arrested cells to enter S phase. In addition, ATM activation by mimosine treatment is mediated by reactive oxygen species (ROS). These results suggest that, upon mimosine treatment, ATM blocks S phase entry in response to ROS, which prevents replication fork stalling-induced DNA damage.
Collapse
Affiliation(s)
- Shoichi Kubota
- From the Department of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hypoxia-induced signaling and its relevance in discovering biomarkers for cancer research. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.bgm.2013.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
47
|
Crocker CD, Chapman LJ, Martínez ML. Hypoxia-induced plasticity in the metabolic response of a widespread cichlid. Comp Biochem Physiol B Biochem Mol Biol 2013; 166:141-7. [PMID: 23973608 DOI: 10.1016/j.cbpb.2013.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/08/2013] [Accepted: 08/15/2013] [Indexed: 10/26/2022]
Abstract
The African cichlid, Pseudocrenilabrus multicolor victoriae is a eurytopic fish that exhibits high levels of developmental plasticity in response to dissolved oxygen availability. In this study, F1 offspring from three sites in the Mpanga River drainage of Western Uganda characterized by different dissolved oxygen (D.O.) regimes were reared under normoxic or hypoxic conditions. After 1 year, enzymes were measured to determine the tissue metabolic capacity of four different tissues: muscle, heart, brain and liver. The enzymes measured were pyruvate kinase [PK], lactate dehydrogenase [LDH], citrate synthase [CS], and cytochrome C oxidase [CCO], and an additional two, malate dehydrogenase (MDH) and fructose 1,6-bisphosphatase (FBPase), were examined in the liver only. Individuals reared under hypoxia exhibited elevated levels of LDH and CCO in the heart; and depressed activity levels of brain CS and liver CCO and MDH relative to normoxia-reared sibs. Results from this study demonstrate that long-term exposure to hypoxia during development can induce changes in the metabolic capacities of P. multicolor. This flexibility may be important in facilitating persistence in variable and/or novel environments, and in the face of increasing global hypoxia.
Collapse
Affiliation(s)
- Cindy D Crocker
- Department of Biology, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
| | | | | |
Collapse
|
48
|
Capitanio JF, Mazza E, Motta M, Mortini P, Reni M. Mechanisms, indications and results of salvage systemic therapy for sporadic and von Hippel–Lindau related hemangioblastomas of the central nervous system. Crit Rev Oncol Hematol 2013; 86:69-84. [DOI: 10.1016/j.critrevonc.2012.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 09/20/2012] [Accepted: 10/11/2012] [Indexed: 10/27/2022] Open
|
49
|
Cui Y, Kim SH, Kim H, Yeom J, Ko K, Park W, Park S. AFM probing the mechanism of synergistic effects of the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) with cefotaxime against extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli. PLoS One 2012; 7:e48880. [PMID: 23152812 PMCID: PMC3496731 DOI: 10.1371/journal.pone.0048880] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 10/01/2012] [Indexed: 11/26/2022] Open
Abstract
Background Extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae poses serious challenges to clinicians because of its resistance to many classes of antibiotics. Methods and Findings The mechanism of synergistic activity of a combination of (−)-epigallocatechin-3-gallate (EGCG) and β-lactam antibiotics cefotaxime was studied on Extended-spectrum β-lactamase producing Escherichia coli (ESBL-EC), by visualizing the morphological alteration on the cell wall induced by the combination using atomic force microscopy (AFM). Cells at sub-MICs (sub-minimum inhibitory concentrations) of cefotaxime were initially filamentated but recovered to the normal shape later, whereas cells at sub-MICs of EGCG experienced temporal disturbance on the cell wall such as leakage and release of cellular debris and groove formation, but later recovered to the normal shape. In contrast, the combination of cefotaxime and EGCG at their respective sub-MICs induced permanent cellular damages as well as continuous elongation in cells and eventually killed them. Flow cytometry showed that intracellular oxidative stress levels in the cell treated with a combination of EGCG and cefotaxime at sub-MICs were higher than those in the cells treated with either cefotaxime or EGCG at sub-MICs. Conclusions These results suggest that the synergistic effect of EGCG between EGCG and cefotaxime against ESBL-EC is related to cooperative activity of exogenous and endogenous reactive oxygen species (ROS) generated by EGCG and cefotaxime, respectively.
Collapse
Affiliation(s)
- Yidan Cui
- Department of Chemistry and Nano Sciences, Ewha Womans University, Seoul, Korea
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - So Hyun Kim
- Department of Chemistry and Nano Sciences, Ewha Womans University, Seoul, Korea
| | - Hyunseok Kim
- Department of Chemistry and Nano Sciences, Ewha Womans University, Seoul, Korea
| | - Jinki Yeom
- Division of Environmental Science and Ecological Engineering, Korea University, Seoul, Korea
| | - Kisung Ko
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Woojun Park
- Division of Environmental Science and Ecological Engineering, Korea University, Seoul, Korea
| | - Sungsu Park
- Department of Chemistry and Nano Sciences, Ewha Womans University, Seoul, Korea
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
50
|
Zhao DJ, Zhang ZY, Harrison J, Kang L. Genome-wide analysis of transcriptional changes in the thoracic muscle of the migratory locust, Locusta migratoria, exposed to hypobaric hypoxia. JOURNAL OF INSECT PHYSIOLOGY 2012; 58:1424-1431. [PMID: 22985864 DOI: 10.1016/j.jinsphys.2012.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 08/10/2012] [Accepted: 08/16/2012] [Indexed: 06/01/2023]
Abstract
Hypobaric hypoxia has both beneficial and detrimental effects on living organisms in high altitude regions. The impact of hypobaric hypoxia has been investigated in numerous vertebrates. However, it is still not well characterized how invertebrates respond to hypobaric hypoxia. In this study, we examined the transcriptional profiles of locust thoracic muscles using microarrays to disclose their strategies to cope with hypobaric hypoxia. We found that hypoxia-inducible factor (HIF) and its target genes did not respond significantly to hypobaric hypoxia. As with severe, normobaric hypoxia, mitochondrial activities were systemically suppressed, mainly involving in energy production and mitochondrial biogenesis. The surveillance processes, involving in clearance of dysfunctional proteins in endoplasmic reticulum, were activated, e.g. endoplasmic reticulum-associated degradation, protein glycosylation, and protein folding. In contrast to severe, normobaric hypoxia, glycolysis was suppressed and the pentose phosphate pathway strengthened. Our data suggested that hypobaric hypoxia induced an oxidative stress rather than an energy crisis in locust thoracic muscles. Our research provides a different perspective of biological responses to hypoxia, complementing the well-studied biological responses to extreme, normobaric hypoxia.
Collapse
Affiliation(s)
- De Jian Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|