51
|
Meng D, Zhang P, Zhang L, Wang H, Ho CT, Li S, Shahidi F, Zhao H. Detection of cellular redox reactions and antioxidant activity assays. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
52
|
Derakhshesh N, Movahedinia A, Salamat N, Hashemitabar M, Bayati V. Using a liver cell culture from Epinephelus coioides as a model to evaluate the nonylphenol-induced oxidative stress. MARINE POLLUTION BULLETIN 2017; 122:243-252. [PMID: 28676171 DOI: 10.1016/j.marpolbul.2017.06.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 06/16/2017] [Accepted: 06/17/2017] [Indexed: 06/07/2023]
Abstract
The present study aimed to use primary liver cell culture derived from the orange-spotted grouper, Epinephelus coioides, to assess the toxic effects of nonylphenol (NP) on the hepatocyte viability and the liver antioxidant system. E. coioides was selected due to its commercial importance. NP was used in this study because of its high potential of producing oxidative stress due to increased reactive oxygen species (ROS). A liver of E. coioides was digested with PBS containing 0.1% collagenase IV. The digested cells were moved to Leibovitz L-15 culture medium with 20% fetal bovine serum (FBS), 100IUmL-1 penicillin, 100μgmL-1 streptomycin. Aliquots of cell suspension were seeded as a monolayer into sterile 25cm2 tissue culture flasks and incubated at 30°C for 14days. The medium, containing non-attached cells, was removed after 24 to 48h and a new medium was added. The IC50 of 10-4molL-1 was determined for nonylphenol using MTT assay. Cells were then incubated with L-15 medium containing 10-5, 2×10-5, 3×10-5molL-1 of NP and samples were taken after 6, 12 and 24h of incubation for analysis of LPO, SOD, CAT, GPx, LDH, AST, ALT, and ALP. Based on the results, the lowest concentration of NP was not markedly cytotoxic to primary hepatocytes and the cell sensitivity to NP increased dose-dependently. The activities of SOD, CAT and GPx decreased significantly, while activities of LPO, LDH, AST, ALT and ALP, increased significantly in a dose-related pattern in NP-treated cells. In conclusion, this study revealed that NP could induce the oxidative stress in cultivated hepatocytes of E. coioides during a short-term exposure. NP toxicity is mainly due to the induction of the reactive oxygen species (ROS), which lead to cell membrane disruption, damage of cellular metabolism, and interference with cellular macromolecules.
Collapse
Affiliation(s)
- Negin Derakhshesh
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, Iran
| | - AbdolAli Movahedinia
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, Iran.
| | - Negin Salamat
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, Iran.
| | - Mahmoud Hashemitabar
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
53
|
Wong BW, Marsch E, Treps L, Baes M, Carmeliet P. Endothelial cell metabolism in health and disease: impact of hypoxia. EMBO J 2017. [PMID: 28637793 DOI: 10.15252/embj.201696150] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In contrast to the general belief, endothelial cell (EC) metabolism has recently been identified as a driver rather than a bystander effect of angiogenesis in health and disease. Indeed, different EC subtypes present with distinct metabolic properties, which determine their function in angiogenesis upon growth factor stimulation. One of the main stimulators of angiogenesis is hypoxia, frequently observed in disease settings such as cancer and atherosclerosis. It has long been established that hypoxic signalling and metabolism changes are highly interlinked. In this review, we will provide an overview of the literature and recent findings on hypoxia-driven EC function and metabolism in health and disease. We summarize evidence on metabolic crosstalk between different hypoxic cell types with ECs and suggest new metabolic targets.
Collapse
Affiliation(s)
- Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Elke Marsch
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Myriam Baes
- Laboratory for Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium .,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
54
|
Bierhansl L, Conradi LC, Treps L, Dewerchin M, Carmeliet P. Central Role of Metabolism in Endothelial Cell Function and Vascular Disease. Physiology (Bethesda) 2017; 32:126-140. [PMID: 28202623 PMCID: PMC5337830 DOI: 10.1152/physiol.00031.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The importance of endothelial cell (EC) metabolism and its regulatory role in the angiogenic behavior of ECs during vessel formation and in the function of different EC subtypes determined by different vascular beds has been recognized only in the last few years. Even more importantly, apart from a role of nitric oxide and reactive oxygen species in EC dysfunction, deregulations of EC metabolism in disease only recently received increasing attention. Although comprehensive metabolic characterization of ECs still needs further investigation, the concept of targeting EC metabolism to treat vascular disease is emerging. In this overview, we summarize EC-specific metabolic pathways, describe the current knowledge on their deregulation in vascular diseases, and give an outlook on how vascular endothelial metabolism can serve as a target to normalize deregulated endothelium.
Collapse
Affiliation(s)
- Laura Bierhansl
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| |
Collapse
|
55
|
Bonfill-Teixidor E, Otxoa-de-Amezaga A, Font-Nieves M, Sans-Fons MG, Planas AM. Differential expression of E-type prostanoid receptors 2 and 4 in microglia stimulated with lipopolysaccharide. J Neuroinflammation 2017; 14:3. [PMID: 28086956 PMCID: PMC5234110 DOI: 10.1186/s12974-016-0780-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/15/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cyclooxygenase-2 (COX-2) is induced under inflammatory conditions, and prostaglandin E2 (PGE2) is one of the products of COX activity. PGE2 has pleiotropic actions depending on the activation of specific E-type prostanoid EP1-4 receptors. We investigated the involvement of PGE2 and EP receptors in glial activation in response to an inflammatory challenge induced by LPS. METHODS Cultures of mouse microglia or astroglia cells were treated with LPS in the presence or absence of COX-2 inhibitors, and the production of PGE2 was measured by ELISA. Cells were treated with PGE2, and the effect on LPS-induced expression of TNF-α messenger RNA (mRNA) and protein was studied in the presence or absence of drug antagonists of the four EP receptors. EP receptor expression and the effects of EP2 and EP4 agonists and antagonists were studied at different time points after LPS. RESULTS PGE2 production after LPS was COX-2-dependent. PGE2 reduced the glial production of TNF-α after LPS. Microglia expressed higher levels of EP4 and EP2 mRNA than astroglia. Activation of EP4 or EP2 receptors with selective drug agonists attenuated LPS-induced TNF-α in microglia. However, only antagonizing EP4 prevented the PGE2 effect demonstrating that EP4 was the main target of PGE2 in naïve microglia. Moreover, the relative expression of EP receptors changed during the course of classical microglial activation since EP4 expression was strongly depressed while EP2 increased 24 h after LPS and was detected in nuclear/peri-nuclear locations. EP2 regulated the expression of iNOS, NADPH oxidase-2, and vascular endothelial growth factor. NADPH oxidase-2 and iNOS activities require the oxidation of NADPH, and the pentose phosphate pathway is a main source of NADPH. LPS increased the mRNA expression of the rate-limiting enzyme of the pentose pathway glucose-6-phosphate dehydrogenase, and EP2 activity was involved in this effect. CONCLUSIONS These results show that while selective activation of EP4 or EP2 exerts anti-inflammatory actions, EP4 is the main target of PGE2 in naïve microglia. The level of EP receptor expression changes from naïve to primed microglia where the COX-2/PGE2/EP2 axis modulates important adaptive metabolic changes.
Collapse
Affiliation(s)
- Ester Bonfill-Teixidor
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Amaia Otxoa-de-Amezaga
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Font-Nieves
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - M Glòria Sans-Fons
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna M Planas
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Rosselló 161 planta 6, 08036, Barcelona, Spain.
| |
Collapse
|
56
|
Hyperlipidemia-Mediated Increased Advanced Lipoxidation End Products Formation, an Important Factor Associated with Decreased Erythrocyte Glucose-6-Phosphate Dehydrogenase Activity in Mild Nonproliferative Diabetic Retinopathy. Can J Diabetes 2016; 41:82-89. [PMID: 27916496 DOI: 10.1016/j.jcjd.2016.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 06/16/2016] [Accepted: 07/25/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVES The present study aimed to evaluate the role of hyperlipidemia in increased formation of advanced lipoxidation end products (ALEs) and to evaluate whether there is any relationship between ALEs generation and erythrocyte glucose-6-phosphate dehydrogenase (G6PD) activity in cases of mild nonproliferative diabetic retinopathy (MNPDR). METHODS In this study, we enrolled 100 patients with type 2 diabetes and MNPDR, 100 subjects with type 2 diabetes but without retinopathy (DNR) and 90 normal individuals without diabetes as healthy controls (HCs). Erythrocyte nicotinamide dinucleotide phosphate (NADPH), G6PD activity, serum total cholesterol, low- and high-density lipoprotein (LDL, HDL) and triglyceride levels were determined by photometric assay. Serum malondialdehyde (MDA) protein adduct and hexanoyl-lysine (HEL) were measured by an enzyme-linked immunosorbent assay (ELISA). RESULTS A robust linear relationship was observed between MDA protein adduct and LDL or cholesterol or triglyceride levels, and HEL and LDL or cholesterol or triglyceride levels in subjects with MNPDR (p=0.0001). A significant inverse association was observed between erythrocyte G6PD activity and serum MDA protein adductor HEL levels in subjects with MNPDR (p=0.0001). CONCLUSIONS Hyperlipidemia is an important factor that is associated with increased ALEs formation in persons with MNPDR. Increased ALEs generation was associated with decreased G6PD activity and low NADPH levels in cases of MNPDR, suggesting their detrimental role in the occurrence of early NPDR.
Collapse
|
57
|
Yang HC, Wu YH, Liu HY, Stern A, Chiu DTY. What has passed is prolog: new cellular and physiological roles of G6PD. Free Radic Res 2016; 50:1047-1064. [PMID: 27684214 DOI: 10.1080/10715762.2016.1223296] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
G6PD deficiency has been the most pervasive inherited disorder in the world since having been discovered. G6PD has an antioxidant role by functioning as a major nicotinamide adenine dinucleotide phosphate (NADPH) provider to reduce excessive oxidative stress. NADPH can produce reactive oxygen species (ROS) and reactive nitrogen species (RNS) mediated by NADPH oxidase (NOX) and nitric oxide synthase (NOS), respectively. Hence, G6PD also has a pro-oxidant role. Research in the past has focused on the enhanced susceptibility of G6PD-deficient cells or individuals to oxidative challenge. The cytoregulatory role of G6PD has largely been overlooked. By using a metabolomic approach, it is noted that upon oxidant challenge, G6PD-deficient cells will reprogram the GSH metabolism from regeneration to synthesis with exhaustive energy consumption. Recently, new cellular/physiologic roles of G6PD have been discovered. By using a proteomic approach, it has been found that G6PD plays a regulatory role in xenobiotic metabolism possibly via NOX and the redox-sensitive Nrf2-signaling pathway to modulate the expression of xenobiotic-metabolizing enzymes. Since G6PD is a key regulator responsible for intracellular redox homeostasis, G6PD deficiency can alter redox balance leading to many abnormal cellular effects such as the cellular inflammatory and immune response against viral infection. G6PD may play an important role in embryogenesis as G6PD-knockdown mouse cannot produce offspring and G6PD-deficient C. elegans with defective egg production and hatching. This array of findings indicates that the cellular and physiologic roles of G6PD, other than the classical role as an antioxidant enzyme, deserve further attention.
Collapse
Affiliation(s)
- Hung-Chi Yang
- a Department of Medical Biotechnology and Laboratory Sciences , College of Medicine, Chang Gung University , Taoyuan , Taiwan.,b Healthy Aging Research Center, Chang Gung University , Taoyuan , Taiwan
| | - Yi-Hsuan Wu
- a Department of Medical Biotechnology and Laboratory Sciences , College of Medicine, Chang Gung University , Taoyuan , Taiwan
| | - Hui-Ya Liu
- a Department of Medical Biotechnology and Laboratory Sciences , College of Medicine, Chang Gung University , Taoyuan , Taiwan
| | - Arnold Stern
- c Department of Biochemistry and Molecular Pharmacology , New York University School of Medicine , New York , NY , USA
| | - Daniel Tsun-Yee Chiu
- a Department of Medical Biotechnology and Laboratory Sciences , College of Medicine, Chang Gung University , Taoyuan , Taiwan.,b Healthy Aging Research Center, Chang Gung University , Taoyuan , Taiwan.,d Department of Pediatric Hematology/Oncology , Chang Gung Memorial Hospital , Linkou , Taiwan
| |
Collapse
|
58
|
Maron BA, Leopold JA. Systems biology: An emerging strategy for discovering novel pathogenetic mechanisms that promote cardiovascular disease. Glob Cardiol Sci Pract 2016; 2016:e201627. [PMID: 29043273 PMCID: PMC5642838 DOI: 10.21542/gcsp.2016.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reductionist theory proposes that analyzing complex systems according to their most fundamental components is required for problem resolution, and has served as the cornerstone of scientific methodology for more than four centuries. However, technological gains in the current scientific era now allow for the generation of large datasets that profile the proteomic, genomic, and metabolomic signatures of biological systems across a range of conditions. The accessibility of data on such a vast scale has, in turn, highlighted the limitations of reductionism, which is not conducive to analyses that consider multiple and contemporaneous interactions between intermediates within a pathway or across constructs. Systems biology has emerged as an alternative approach to analyze complex biological systems. This methodology is based on the generation of scale-free networks and, thus, provides a quantitative assessment of relationships between multiple intermediates, such as protein-protein interactions, within and between pathways of interest. In this way, systems biology is well positioned to identify novel targets implicated in the pathogenesis or treatment of diseases. In this review, the historical root and fundamental basis of systems biology, as well as the potential applications of this methodology are discussed with particular emphasis on integration of these concepts to further understanding of cardiovascular disorders such as coronary artery disease and pulmonary hypertension.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
59
|
Ham M, Choe SS, Shin KC, Choi G, Kim JW, Noh JR, Kim YH, Ryu JW, Yoon KH, Lee CH, Kim JB. Glucose-6-Phosphate Dehydrogenase Deficiency Improves Insulin Resistance With Reduced Adipose Tissue Inflammation in Obesity. Diabetes 2016; 65:2624-38. [PMID: 27284106 DOI: 10.2337/db16-0060] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/24/2016] [Indexed: 11/13/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD), a rate-limiting enzyme of the pentose phosphate pathway, plays important roles in redox regulation and de novo lipogenesis. It was recently demonstrated that aberrant upregulation of G6PD in obese adipose tissue mediates insulin resistance as a result of imbalanced energy metabolism and oxidative stress. It remains elusive, however, whether inhibition of G6PD in vivo may relieve obesity-induced insulin resistance. In this study we showed that a hematopoietic G6PD defect alleviates insulin resistance in obesity, accompanied by reduced adipose tissue inflammation. Compared with wild-type littermates, G6PD-deficient mutant (G6PD(mut)) mice were glucose tolerant upon high-fat-diet (HFD) feeding. Intriguingly, the expression of NADPH oxidase genes to produce reactive oxygen species was alleviated, whereas that of antioxidant genes was enhanced in the adipose tissue of HFD-fed G6PD(mut) mice. In diet-induced obesity (DIO), the adipose tissue of G6PD(mut) mice decreased the expression of inflammatory cytokines, accompanied by downregulated proinflammatory macrophages. Accordingly, macrophages from G6PD(mut) mice greatly suppressed lipopolysaccharide-induced proinflammatory signaling cascades, leading to enhanced insulin sensitivity in adipocytes and hepatocytes. Furthermore, adoptive transfer of G6PD(mut) bone marrow to wild-type mice attenuated adipose tissue inflammation and improved glucose tolerance in DIO. Collectively, these data suggest that inhibition of macrophage G6PD would ameliorate insulin resistance in obesity through suppression of proinflammatory responses.
Collapse
Affiliation(s)
- Mira Ham
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Sung Sik Choe
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Kyung Cheul Shin
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Goun Choi
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| | - Ji-Won Kim
- Department of Endocrinology and Metabolism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, Korea
| | - Je-Won Ryu
- Department of Radiation Oncology, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Korea
| | - Kun-Ho Yoon
- Department of Endocrinology and Metabolism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon, Korea
| | - Jae Bum Kim
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, National Creative Research Initiatives Center for Adipose Tissue Remodeling, Seoul National University, Seoul, Korea
| |
Collapse
|
60
|
Pircher A, Treps L, Bodrug N, Carmeliet P. Endothelial cell metabolism: A novel player in atherosclerosis? Basic principles and therapeutic opportunities. Atherosclerosis 2016; 253:247-257. [PMID: 27594537 DOI: 10.1016/j.atherosclerosis.2016.08.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/09/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022]
Abstract
Atherosclerosis is a leading cause of morbidity and mortality in Western society. Despite improved insight into disease pathogenesis and therapeutic options, additional treatment strategies are required. Emerging evidence highlights the relevance of endothelial cell (EC) metabolism for angiogenesis, and indicates that EC metabolism is perturbed when ECs become dysfunctional to promote atherogenesis. In this review, we overview the latest insights on EC metabolism and discuss current knowledge on how atherosclerosis deregulates EC metabolism, and how maladaptation of deregulated EC metabolism can contribute to atherosclerosis progression. We will also highlight possible therapeutic avenues, based on targeting EC metabolism.
Collapse
Affiliation(s)
- Andreas Pircher
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium
| | - Natalia Bodrug
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium; Laboratory of Adhesion and Angiogenesis, Centre for Tumour Biology, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, B-3000, Belgium.
| |
Collapse
|
61
|
Treps L, Conradi LC, Harjes U, Carmeliet P. Manipulating Angiogenesis by Targeting Endothelial Metabolism: Hitting the Engine Rather than the Drivers—A New Perspective? Pharmacol Rev 2016; 68:872-87. [DOI: 10.1124/pr.116.012492] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
62
|
Shah SS, Rockett KA, Jallow M, Sisay-Joof F, Bojang KA, Pinder M, Jeffreys A, Craik R, Hubbart C, Wellems TE, Kwiatkowski DP. Heterogeneous alleles comprising G6PD deficiency trait in West Africa exert contrasting effects on two major clinical presentations of severe malaria. Malar J 2016; 15:13. [PMID: 26738565 PMCID: PMC4704392 DOI: 10.1186/s12936-015-1045-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/09/2015] [Indexed: 11/11/2022] Open
Abstract
Background Glucose-6-phosphate dehydrogenase (G6PD) deficiency exhibits considerable allelic heterogeneity which manifests with variable biochemical and clinical penetrance. It has long been thought that G6PD deficiency confers partial protection against severe malaria, however prior genetic association studies have disagreed with regard to the strength and specificity of a protective effect, which might reflect differences in the host genetic background, environmental influences, or in the specific clinical phenotypes considered. Methods A case-control association study of severe malaria was conducted in The Gambia, a region in West Africa where there is considerable allelic heterogeneity underlying expression of G6PD deficiency trait, evaluating the three major nonsynonymous polymorphisms known to be associated with enzyme deficiency (A968G, T542A, and C202T) in a cohort of 3836 controls and 2379 severe malaria cases. Results Each deficiency allele exhibited a similar trend toward protection against severe malaria overall (15–26 % reduced risk); however, in stratifying severe malaria to two of its constituent clinical subphenotypes, severe malarial anaemia (SMA) and cerebral malaria (CM), the three deficiency alleles exhibited trends of opposing effect, with risk conferred to SMA and protection with respect to CM. To assess the overall effect of G6PD deficiency trait, deficiency alleles found across all three loci were pooled. G6PD deficiency trait was found to be significantly associated with protection from severe malaria overall (OR 0.83 [0.75–0.92], \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 0.0006$$\end{document}P=0.0006), but this was limited to CM (OR 0.73 [0.61–0.87], \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 0.0005$$\end{document}P=0.0005), with a trend toward increased risk for SMA, especially in fully-deficient individuals (OR 1.43 [0.99–2.08], \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 0.056$$\end{document}P=0.056). Sex-stratified testing largely comported with these results, with evidence suggesting that protection by G6PD deficiency trait is conferred to both males and females, though susceptibility to SMA may be restricted to fully-deficient male hemizygotes. Conclusions In a part of Africa where multiple alleles contribute to expression of G6PD deficiency trait, these findings clarify and extend previous work done in populations where a single variant predominates, and taken together suggest a causal role for G6PD deficiency trait itself with respect to severe malaria, with opposing effects seen on two major clinical subphenotypes.
Collapse
Affiliation(s)
- Shivang S Shah
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. .,Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Kirk A Rockett
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Muminatou Jallow
- Medical Research Council Laboratories, Banjul, Fajara, The Gambia.
| | - Fatou Sisay-Joof
- Medical Research Council Laboratories, Banjul, Fajara, The Gambia.
| | - Kalifa A Bojang
- Medical Research Council Laboratories, Banjul, Fajara, The Gambia.
| | - Margaret Pinder
- Medical Research Council Laboratories, Banjul, Fajara, The Gambia.
| | - Anna Jeffreys
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Rachel Craik
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Christina Hubbart
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Dominic P Kwiatkowski
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. .,Wellcome Trust Sanger Institute, Hinxton, UK.
| | | |
Collapse
|
63
|
Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov 2015; 15:125-42. [PMID: 26612664 DOI: 10.1038/nrd.2015.2] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulation of vascular permeability, recruitment of leukocytes from blood to tissue and angiogenesis are all processes that occur at the level of the microvasculature during both physiological and pathological conditions. The interplay between microvascular cells and leukocytes during inflammation, together with the emerging roles of leukocytes in the modulation of the angiogenic process, make leukocyte-vascular interactions prime targets for therapeutics to potentially treat a wide range of diseases, including pathological and dysfunctional vessel growth, chronic inflammation and fibrosis. In this Review, we discuss how the different cell types that are present in and around microvessels interact, cooperate and instruct each other, and in this context we highlight drug targets as well as emerging druggable processes that can be exploited to restore tissue homeostasis.
Collapse
Affiliation(s)
- Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| |
Collapse
|
64
|
Smith GA, Fearnley GW, Harrison MA, Tomlinson DC, Wheatcroft SB, Ponnambalam S. Vascular endothelial growth factors: multitasking functionality in metabolism, health and disease. J Inherit Metab Dis 2015; 38:753-63. [PMID: 25868665 DOI: 10.1007/s10545-015-9838-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/06/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
Vascular endothelial growth factors (VEGFs) bind to VEGF receptor tyrosine kinases (VEGFRs). The VEGF and VEGFR gene products regulate diverse regulatory pathways in mammalian development, health and disease. The interaction between a particular VEGF and its cognate VEGFR activates multiple signal transduction pathways which regulate different cellular responses including metabolism, gene expression, proliferation, migration, and survival. The family of VEGF isoforms regulate vascular physiology and promote tissue homeostasis. VEGF dysfunction is implicated in major chronic disease states including atherosclerosis, diabetes, and cancer. More recent studies implicate a strong link between response to VEGF and regulation of vascular metabolism. Understanding how this family of multitasking cytokines regulates cell and animal function has implications for treating many different diseases.
Collapse
Affiliation(s)
- Gina A Smith
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | | | | | | | | | |
Collapse
|
65
|
|
66
|
Verdegem D, Moens S, Stapor P, Carmeliet P. Endothelial cell metabolism: parallels and divergences with cancer cell metabolism. Cancer Metab 2014; 2:19. [PMID: 25250177 PMCID: PMC4171726 DOI: 10.1186/2049-3002-2-19] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 02/08/2023] Open
Abstract
The stromal vasculature in tumors is a vital conduit of nutrients and oxygen for cancer cells. To date, the vast majority of studies have focused on unraveling the genetic basis of vessel sprouting (also termed angiogenesis). In contrast to the widely studied changes in cancer cell metabolism, insight in the metabolic regulation of angiogenesis is only just emerging. These studies show that metabolic pathways in endothelial cells (ECs) importantly regulate angiogenesis in conjunction with genetic signals. In this review, we will highlight these emerging insights in EC metabolism and discuss them in perspective of cancer cell metabolism. While it is generally assumed that cancer cells have unique metabolic adaptations, not shared by healthy non-transformed cells, we will discuss parallels and highlight differences between endothelial and cancer cell metabolism and consider possible novel therapeutic opportunities arising from targeting both cancer and endothelial cells.
Collapse
Affiliation(s)
- Dries Verdegem
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| |
Collapse
|
67
|
Stapor P, Wang X, Goveia J, Moens S, Carmeliet P. Angiogenesis revisited - role and therapeutic potential of targeting endothelial metabolism. J Cell Sci 2014; 127:4331-41. [PMID: 25179598 DOI: 10.1242/jcs.153908] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Clinically approved therapies that target angiogenesis in tumors and ocular diseases focus on controlling pro-angiogenic growth factors in order to reduce aberrant microvascular growth. Although research on angiogenesis has revealed key mechanisms that regulate tissue vascularization, therapeutic success has been limited owing to insufficient efficacy, refractoriness and tumor resistance. Emerging concepts suggest that, in addition to growth factors, vascular metabolism also regulates angiogenesis and is a viable target for manipulating the microvasculature. Recent studies show that endothelial cells rely on glycolysis for ATP production, and that the key glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) regulates angiogenesis by controlling the balance of tip versus stalk cells. As endothelial cells acquire a tip cell phenotype, they increase glycolytic production of ATP for sprouting. Furthermore, pharmacological blockade of PFKFB3 causes a transient, partial reduction in glycolysis, and reduces pathological angiogenesis with minimal systemic harm. Although further assessment of endothelial cell metabolism is necessary, these results represent a paradigm shift in anti-angiogenic therapy from targeting angiogenic factors to focusing on vascular metabolism, warranting research on the metabolic pathways that govern angiogenesis.
Collapse
Affiliation(s)
- Peter Stapor
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Xingwu Wang
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, B-3000 Leuven, Belgium Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
68
|
Ho HY, Cheng ML, Chiu DTY. Glucose-6-phosphate dehydrogenase--beyond the realm of red cell biology. Free Radic Res 2014; 48:1028-48. [PMID: 24720642 DOI: 10.3109/10715762.2014.913788] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is critical to the maintenance of NADPH pool and redox homeostasis. Conventionally, G6PD deficiency has been associated with hemolytic disorders. Most biochemical variants were identified and characterized at molecular level. Recently, a number of studies have shone light on the roles of G6PD in aspects of physiology other than erythrocytic pathophysiology. G6PD deficiency alters the redox homeostasis, and affects dysfunctional cell growth and signaling, anomalous embryonic development, and altered susceptibility to infection. The present article gives a brief review of basic science and clinical findings about G6PD, and covers the latest development in the field. Moreover, how G6PD status alters the susceptibility of the affected individuals to certain degenerative diseases is also discussed.
Collapse
Affiliation(s)
- H-Y Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University , Kwei-san, Tao-yuan , Taiwan
| | | | | |
Collapse
|
69
|
Xu Y, An X, Guo X, Habtetsion TG, Wang Y, Xu X, Kandala S, Li Q, Li H, Zhang C, Caldwell RB, Fulton DJ, Su Y, Hoda MN, Zhou G, Wu C, Huo Y. Endothelial PFKFB3 plays a critical role in angiogenesis. Arterioscler Thromb Vasc Biol 2014; 34:1231-9. [PMID: 24700124 DOI: 10.1161/atvbaha.113.303041] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular cells, particularly endothelial cells, adopt aerobic glycolysis to generate energy to support cellular functions. The effect of endothelial glycolysis on angiogenesis remains unclear. 6-Phosphofructo-2-kinase/fructose-2, 6-bisphosphatase, isoform 3 (PFKFB3) is a critical enzyme for endothelial glycolysis. By blocking or deleting PFKFB3 in endothelial cells, we investigated the influence of endothelial glycolysis on angiogenesis both in vitro and in vivo. APPROACH AND RESULTS Under hypoxic conditions or after treatment with angiogenic factors, endothelial PFKFB3 was upregulated both in vitro and in vivo. The knockdown or overexpression of PFKFB3 suppressed or accelerated endothelial proliferation and migration in vitro, respectively. Neonatal mice from a model of oxygen-induced retinopathy showed suppressed neovascular growth in the retina when endothelial PFKFB3 was genetically deleted or when the mice were treated with a PFKFB3 inhibitor. In addition, tumors implanted in mice deficient in endothelial PFKFB3 grew more slowly and were provided with less blood flow. A lower level of phosphorylated protein kinase B was observed in PFKFB3-knockdown endothelial cells, which was accompanied by a decrease in intracellular lactate. The addition of lactate to PFKFB3-knockdown cells rescued the suppression of endothelial proliferation and migration. CONCLUSIONS The blockade or deletion of endothelial PFKFB3 decreases angiogenesis both in vitro and in vivo. Thus, PFKFB3 is a promising target for the reduction of endothelial glycolysis and its related pathological angiogenesis.
Collapse
Affiliation(s)
- Yiming Xu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xiaofei An
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xin Guo
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Tsadik Ghebreamlak Habtetsion
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Yong Wang
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xizhen Xu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Sridhar Kandala
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Qinkai Li
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Honggui Li
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Chunxiang Zhang
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Ruth B Caldwell
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - David J Fulton
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Yunchao Su
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Md Nasrul Hoda
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Gang Zhou
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Chaodong Wu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.).
| | - Yuqing Huo
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.).
| |
Collapse
|
70
|
Schoors S, De Bock K, Cantelmo AR, Georgiadou M, Ghesquière B, Cauwenberghs S, Kuchnio A, Wong BW, Quaegebeur A, Goveia J, Bifari F, Wang X, Blanco R, Tembuyser B, Cornelissen I, Bouché A, Vinckier S, Diaz-Moralli S, Gerhardt H, Telang S, Cascante M, Chesney J, Dewerchin M, Carmeliet P. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis. Cell Metab 2014; 19:37-48. [PMID: 24332967 DOI: 10.1016/j.cmet.2013.11.008] [Citation(s) in RCA: 402] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 01/26/2023]
Abstract
Strategies targeting pathological angiogenesis have focused primarily on blocking vascular endothelial growth factor (VEGF), but resistance and insufficient efficacy limit their success, mandating alternative antiangiogenic strategies. We recently provided genetic evidence that the glycolytic activator phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) promotes vessel formation but did not explore the antiangiogenic therapeutic potential of PFKFB3 blockade. Here, we show that blockade of PFKFB3 by the small molecule 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) reduced vessel sprouting in endothelial cell (EC) spheroids, zebrafish embryos, and the postnatal mouse retina by inhibiting EC proliferation and migration. 3PO also suppressed vascular hyperbranching induced by inhibition of Notch or VEGF receptor 1 (VEGFR1) and amplified the antiangiogenic effect of VEGF blockade. Although 3PO reduced glycolysis only partially and transiently in vivo, this sufficed to decrease pathological neovascularization in ocular and inflammatory models. These insights may offer therapeutic antiangiogenic opportunities.
Collapse
Affiliation(s)
- Sandra Schoors
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Katrien De Bock
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Maria Georgiadou
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Bart Ghesquière
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Sandra Cauwenberghs
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Anna Kuchnio
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Annelies Quaegebeur
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Francesco Bifari
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Xingwu Wang
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Raquel Blanco
- Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3LY, UK
| | - Bieke Tembuyser
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Ivo Cornelissen
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Ann Bouché
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Santiago Diaz-Moralli
- Department of Biochemistry and Molecular Biology and IBUB, Universitat de Barcelona, Barcelona 08007, Spain
| | - Holger Gerhardt
- Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3LY, UK; Vascular Patterning Laboratory, Vesalius Research Center, University of Leuven, Leuven 3000, Belgium; Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Sucheta Telang
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Marta Cascante
- Department of Biochemistry and Molecular Biology and IBUB, Universitat de Barcelona, Barcelona 08007, Spain
| | - Jason Chesney
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven 3000, Belgium.
| |
Collapse
|
71
|
Eelen G, Cruys B, Welti J, De Bock K, Carmeliet P. Control of vessel sprouting by genetic and metabolic determinants. Trends Endocrinol Metab 2013; 24:589-96. [PMID: 24075830 DOI: 10.1016/j.tem.2013.08.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/26/2013] [Accepted: 08/28/2013] [Indexed: 01/28/2023]
Abstract
Vessel sprouting by endothelial cells (ECs) during angiogenesis relies on a navigating tip cell and on proliferating stalk cells that elongate the shaft. To date, only genetic signals have been shown to regulate vessel sprouting. However, emerging evidence indicates that the angiogenic switch also requires a metabolic switch. Indeed, angiogenic signals not only induce a change in EC metabolism but this metabolic adaptation also co-determines vessel sprouting. The glycolytic activator PFKFB3 regulates stalk cell proliferation and renders ECs more competitive to reach the tip. We discuss the emerging link between angiogenesis and EC metabolism during the various stages of vessel sprouting, focusing only on genetic signals for which an effect on EC metabolism has been documented.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Department of Oncology, Katholieke Universiteit Leuven (KU Leuven), Herestraat 49, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
72
|
Handy DE, Loscalzo J, Leopold JA. Systems analysis of oxidant stress in the vasculature. IUBMB Life 2013; 65:911-20. [PMID: 24265198 DOI: 10.1002/iub.1221] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/01/2013] [Indexed: 01/11/2023]
Abstract
Systems biology and network analysis are emerging as valuable tools for the discovery of novel relationships, the identification of key regulatory factors, and the prediction of phenotypic changes in complex biological systems. Redox homeostasis in the vasculature is maintained by an intricate balance between oxidant-generating and antioxidant systems. When these systems are perturbed, conditions are permissive for oxidant stress, which, in turn, promotes vascular dysfunction and structural remodeling. Owing to the number of elements involved in redox regulation and the different vascular pathophenotypes associated with oxidant stress, vascular oxidant stress represents an ideal system to study by network analysis. Networks offer a method to organize experimentally derived factors, including proteins, metabolites, and DNA, that are represented as nodes into an unbiased comprehensive platform for study. Through analysis of the network, it is possible to determine essential or regulatory nodes, identify previously unknown connections between nodes, and locate modules, which are groups of nodes located within the same neighborhood that function together and have implications for phenotype. Investigators have only recently begun to construct oxidant stress-related networks to examine vascular structure and function; however, these early studies have provided mechanistic insight to further our understanding of this complicated biological system.
Collapse
Affiliation(s)
- Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
73
|
Abstract
Endothelial cells (ECs) are quiescent for years but can plastically switch to angiogenesis. Vascular sprouting relies on the coordinated activity of migrating tip cells at the forefront and proliferating stalk cells that elongate the sprout. Past studies have identified genetic signals that control vascular branching. Prominent are VEGF, activating tip cells, and Notch, which stimulates stalk cells. After the branch is formed and perfused, ECs become quiescent phalanx cells. Now, emerging evidence has accumulated indicating that ECs not only adapt their metabolism when switching from quiescence to sprouting but also that metabolism regulates vascular sprouting in parallel to the control by genetic signals.
Collapse
Affiliation(s)
- Katrien De Bock
- Department of Oncology, University of Leuven, Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Leuven 3000, Belgium; VIB, Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Leuven 3000, Belgium
| | | | | |
Collapse
|
74
|
Macrophage glucose-6-phosphate dehydrogenase stimulates proinflammatory responses with oxidative stress. Mol Cell Biol 2013; 33:2425-35. [PMID: 23572562 DOI: 10.1128/mcb.01260-12] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme that regulates cellular redox potential. In this study, we demonstrate that macrophage G6PD plays an important role in the modulation of proinflammatory responses and oxidative stress. The G6PD levels in macrophages in the adipose tissue of obese animals were elevated, and G6PD mRNA levels positively correlated with those of proinflammatory genes. Lipopolysaccharide (LPS) and free fatty acids, which initiate proinflammatory signals, stimulated macrophage G6PD. Overexpression of macrophage G6PD potentiated the expression of proinflammatory and pro-oxidative genes responsible for the aggravation of insulin sensitivity in adipocytes. In contrast, when macrophage G6PD was inhibited or suppressed via chemical inhibitors or small interfering RNA (siRNA), respectively, basal and LPS-induced proinflammatory gene expression was attenuated. Furthermore, macrophage G6PD increased activation of the p38 mitogen-activated protein kinase (MAPK) and NF-κB pathways, which may lead to a vicious cycle of oxidative stress and proinflammatory cascade. Together, these data suggest that an abnormal increase of G6PD in macrophages promotes oxidative stress and inflammatory responses in the adipose tissue of obese animals.
Collapse
|
75
|
Connes P, Verlhac S, Bernaudin F. Advances in understanding the pathogenesis of cerebrovascular vasculopathy in sickle cell anaemia. Br J Haematol 2013; 161:484-98. [DOI: 10.1111/bjh.12300] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Philippe Connes
- UMR Inserm 665; Academic Hospital of Pointe à Pitre; Pointe à Pitre Guadeloupe
- Laboratory ACTES EA3596; Department of Physiology; University of Antilles-Guyane; Pointe à Pitre Guadeloupe
- Laboratory of Excellence GR-Ex “The red cell: from genesis to death”; PRES Sorbonne Paris Cité; Paris France
| | - Suzanne Verlhac
- Medical Imaging; Centre Hospitalier Intercommunal; Creteil France
| | - Françoise Bernaudin
- Department of Paediatrics; Referral Centre for Sickle Cell Disease; Centre Hospitalier Intercommunal; Creteil France
| |
Collapse
|
76
|
Karagiannis GS, Weile J, Bader GD, Minta J. Integrative pathway dissection of molecular mechanisms of moxLDL-induced vascular smooth muscle phenotype transformation. BMC Cardiovasc Disord 2013; 13:4. [PMID: 23324130 PMCID: PMC3556327 DOI: 10.1186/1471-2261-13-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 12/29/2012] [Indexed: 01/08/2023] Open
Abstract
Background Atherosclerosis (AT) is a chronic inflammatory disease characterized by the accumulation of inflammatory cells, lipoproteins and fibrous tissue in the walls of arteries. AT is the primary cause of heart attacks and stroke and is the leading cause of death in Western countries. To date, the pathogenesis of AT is not well-defined. Studies have shown that the dedifferentiation of contractile and quiescent vascular smooth muscle cells (SMC) to the proliferative, migratory and synthetic phenotype in the intima is pivotal for the onset and progression of AT. To further delineate the mechanisms underlying the pathogenesis of AT, we analyzed the early molecular pathways and networks involved in the SMC phenotype transformation. Methods Quiescent human coronary artery SMCs were treated with minimally-oxidized LDL (moxLDL), for 3 hours and 21 hours, respectively. Transcriptomic data was generated for both time-points using microarrays and was subjected to pathway analysis using Gene Set Enrichment Analysis, GeneMANIA and Ingenuity software tools. Gene expression heat maps and pathways enriched in differentially expressed genes were compared to identify functional biological themes to elucidate early and late molecular mechanisms of moxLDL-induced SMC dedifferentiation. Results Differentially expressed genes were found to be enriched in cholesterol biosynthesis, inflammatory cytokines, chemokines, growth factors, cell cycle control and myogenic contraction themes. These pathways are consistent with inflammatory responses, cell proliferation, migration and ECM production, which are characteristic of SMC dedifferentiation. Furthermore, up-regulation of cholesterol synthesis and dysregulation of cholesterol metabolism was observed in moxLDL-induced SMC. These observations are consistent with the accumulation of cholesterol and oxidized cholesterol esters, which induce proinflammatory reactions during atherogenesis. Our data implicate for the first time IL12, IFN-α, HGF, CSF3, and VEGF signaling in SMC phenotype transformation. GPCR signaling, HBP1 (repressor of cyclin D1 and CDKN1B), and ID2 and ZEB1 transcriptional regulators were also found to have important roles in SMC dedifferentiation. Several microRNAs were observed to regulate the SMC phenotype transformation via an interaction with IFN-γ pathway. Also, several “nexus” genes in complex networks, including components of the multi-subunit enzyme complex involved in the terminal stages of cholesterol synthesis, microRNAs (miR-203, miR-511, miR-590-3p, miR-346*/miR- 1207-5p/miR-4763-3p), GPCR proteins (GPR1, GPR64, GPRC5A, GPR171, GPR176, GPR32, GPR25, GPR124) and signal transduction pathways, were found to be regulated. Conclusions The systems biology analysis of the in vitro model of moxLDL-induced VSMC phenotype transformation was associated with the regulation of several genes not previously implicated in SMC phenotype transformation. The identification of these potential candidate genes enable hypothesis generation and in vivo functional experimentation (such as gain and loss-of-function studies) to establish causality with the process of SMC phenotype transformation and atherogenesis.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, and Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, M5S 1A8, Canada
| | | | | | | |
Collapse
|
77
|
Pinna A, Contini EL, Carru C, Solinas G. Glucose-6-phosphate dehydrogenase deficiency and diabetes mellitus with severe retinal complications in a Sardinian population, Italy. Int J Med Sci 2013; 10:1907-13. [PMID: 24324368 PMCID: PMC3856382 DOI: 10.7150/ijms.6776] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/14/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency is one of the most common human genetic abnormalities, with a high prevalence in Sardinia, Italy. Evidence indicates that G6PD-deficient patients are protected against vascular disease. Little is known about the relationship between G6PD deficiency and diabetes mellitus. The purpose of this study was to compare G6PD deficiency prevalence in Sardinian diabetic men with severe retinal vascular complications and in age-matched non-diabetic controls and ascertain whether G6PD deficiency may offer protection against this vascular disorder. METHODS Erythrocyte G6PD activity was determined using a quantitative assay in 390 diabetic men with proliferative diabetic retinopathy (PDR) and 390 male non-diabetic controls, both aged ≥50 years. Conditional logistic regression models were used to investigate the association between G6PD deficiency and diabetes with severe retinal complications. RESULTS G6PD deficiency was found in 21 (5.4 %) diabetic patients and 33 (8.5 %) controls (P=0.09). In a univariate conditional logistic regression model, G6PD deficiency showed a trend for protection against diabetes with PDR, but the odds ratio (OR) fell short of statistical significance (OR=0.6, 95% confidence interval=0.35-1.08, P=0.09). In multivariate conditional logistic regression models, including as covariates G6PD deficiency, plasma glucose, and systemic hypertension or systolic or diastolic blood pressure, G6PD deficiency showed no statistically significant protection against diabetes with PDR. CONCLUSIONS The prevalence of G6PD deficiency in diabetic men with PDR was lower than in age-matched non-diabetic controls. G6PD deficiency showed a trend for protection against diabetes with PDR, but results were not statistically significant.
Collapse
Affiliation(s)
- Antonio Pinna
- 1. Department of Surgical, Microsurgical, & Medical Sciences, Section of Ophthalmology, University of Sassari, Sassari, Italy
| | | | | | | |
Collapse
|
78
|
Zhang Z, Yang Z, Zhu B, Hu J, Liew CW, Zhang Y, Leopold JA, Handy DE, Loscalzo J, Stanton RC. Increasing glucose 6-phosphate dehydrogenase activity restores redox balance in vascular endothelial cells exposed to high glucose. PLoS One 2012. [PMID: 23185302 PMCID: PMC3501497 DOI: 10.1371/journal.pone.0049128] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previous studies have shown that high glucose increases reactive oxygen species (ROS) in endothelial cells that contributes to vascular dysfunction and atherosclerosis. Accumulation of ROS is due to dysregulated redox balance between ROS-producing systems and antioxidant systems. Previous research from our laboratory has shown that high glucose decreases the principal cellular reductant, NADPH by impairing the activity of glucose 6-phosphate dehydrogenase (G6PD). We and others also have shown that the high glucose-induced decrease in G6PD activity is mediated, at least in part, by cAMP-dependent protein kinase A (PKA). As both the major antioxidant enzymes and NADPH oxidase, a major source of ROS, use NADPH as substrate, we explored whether G6PD activity was a critical mediator of redox balance. We found that overexpression of G6PD by pAD-G6PD infection restored redox balance. Moreover inhibition of PKA decreased ROS accumulation and increased redox enzymes, while not altering the protein expression level of redox enzymes. Interestingly, high glucose stimulated an increase in NADPH oxidase (NOX) and colocalization of G6PD with NOX, which was inhibited by the PKA inhibitor. Lastly, inhibition of PKA ameliorated high glucose mediated increase in cell death and inhibition of cell growth. These studies illustrate that increasing G6PD activity restores redox balance in endothelial cells exposed to high glucose, which is a potentially important therapeutic target to protect ECs from the deleterious effects of high glucose.
Collapse
Affiliation(s)
- Zhaoyun Zhang
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Endocrinology and Metabolism, Huashan Hospital, Shanghai, China
| | - Zhihong Yang
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bo Zhu
- Division of Endocrinology and Metabolism, Nanfang Hospital, Guangzhou, China
| | - Ji Hu
- Division of Endocrinology and Metabolism, 2nd Affiliated Hospital of Soochow University, Suzhou, China
| | - Chong Wee Liew
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yingyi Zhang
- Brigham Woman's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jane A. Leopold
- Brigham Woman's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Diane E. Handy
- Brigham Woman's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joseph Loscalzo
- Brigham Woman's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert C. Stanton
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
79
|
Riganti C, Gazzano E, Polimeni M, Aldieri E, Ghigo D. The pentose phosphate pathway: an antioxidant defense and a crossroad in tumor cell fate. Free Radic Biol Med 2012; 53:421-36. [PMID: 22580150 DOI: 10.1016/j.freeradbiomed.2012.05.006] [Citation(s) in RCA: 308] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 04/14/2012] [Accepted: 05/03/2012] [Indexed: 01/10/2023]
Abstract
The pentose phosphate pathway, one of the main antioxidant cellular defense systems, has been related for a long time almost exclusively to its role as a provider of reducing power and ribose phosphate to the cell. In addition to this "traditional" correlation, in the past years multiple roles have emerged for this metabolic cascade, involving the cell cycle, apoptosis, differentiation, motility, angiogenesis, and the response to anti-tumor therapy. These findings make the pentose phosphate pathway a very interesting target in tumor cells. This review summarizes the latest discoveries relating the activity of the pentose phosphate pathway to various aspects of tumor metabolism, such as cell proliferation and death, tissue invasion, angiogenesis, and resistance to therapy, and discusses the possibility that drugs modulating the pathway could be used as potential tools in tumor therapy.
Collapse
Affiliation(s)
- Chiara Riganti
- Department of Genetics, Biology, and Biochemistry, University of Torino, Turin, Italy.
| | | | | | | | | |
Collapse
|
80
|
Handy DE, Loscalzo J. Redox regulation of mitochondrial function. Antioxid Redox Signal 2012; 16:1323-67. [PMID: 22146081 PMCID: PMC3324814 DOI: 10.1089/ars.2011.4123] [Citation(s) in RCA: 372] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 12/06/2011] [Accepted: 12/06/2011] [Indexed: 02/06/2023]
Abstract
Redox-dependent processes influence most cellular functions, such as differentiation, proliferation, and apoptosis. Mitochondria are at the center of these processes, as mitochondria both generate reactive oxygen species (ROS) that drive redox-sensitive events and respond to ROS-mediated changes in the cellular redox state. In this review, we examine the regulation of cellular ROS, their modes of production and removal, and the redox-sensitive targets that are modified by their flux. In particular, we focus on the actions of redox-sensitive targets that alter mitochondrial function and the role of these redox modifications on metabolism, mitochondrial biogenesis, receptor-mediated signaling, and apoptotic pathways. We also consider the role of mitochondria in modulating these pathways, and discuss how redox-dependent events may contribute to pathobiology by altering mitochondrial function.
Collapse
Affiliation(s)
- Diane E Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
81
|
Stanton RC. Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life 2012; 64:362-9. [PMID: 22431005 DOI: 10.1002/iub.1017] [Citation(s) in RCA: 466] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 02/07/2012] [Indexed: 02/06/2023]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme of the pentose phosphate pathway. Many scientists think that the roles and regulation of G6PD in physiology and pathophysiology have been well established as the enzyme was first identified 80 years ago. And that G6PD has been extensively studied especially with respect to G6PD deficiency and its association with hemolysis, and with respect to the role G6PD plays in lipid metabolism. But there has been a growing understanding of the central importance of G6PD to cellular physiology as it is a major source of NADPH that is required by many essential cellular systems including the antioxidant pathways, nitric oxide synthase, NADPH oxidase, cytochrome p450 system, and others. Indeed G6PD is essential for cell survival. It has also become evident that G6PD is highly regulated by many signals that affect transcription, post-translation, intracellular location, and interactions with other protein. Pathophysiologic roles for G6PD have also been identified in such disease processes as diabetes, aldosterone-induced endothelial dysfunction, cancer, and others. It is now clear that G6PD is under complex regulatory control and of central importance to many cellular processes. In this review the biochemistry, regulatory signals, physiologic roles, and pathophysiologic roles for G6PD that have been elucidated over the past 20 years are discussed.
Collapse
Affiliation(s)
- Robert C Stanton
- Renal Section, Joslin Diabetes Center, and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
82
|
Eynan M, Tsitlovsky D, Batit L, Hochman A, Krinsky N, Abramovich A. Is glucose-6-phosphate dehydrogenase deficiency a risk factor for hyperbaric oxygen exposure? Eur J Appl Physiol 2011; 112:2549-56. [DOI: 10.1007/s00421-011-2229-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 10/27/2011] [Indexed: 10/15/2022]
|
83
|
Cappai G, Songini M, Doria A, Cavallerano JD, Lorenzi M. Increased prevalence of proliferative retinopathy in patients with type 1 diabetes who are deficient in glucose-6-phosphate dehydrogenase. Diabetologia 2011; 54:1539-42. [PMID: 21380594 DOI: 10.1007/s00125-011-2099-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 02/04/2011] [Indexed: 10/18/2022]
Abstract
AIMS/HYPOTHESIS Impaired activity of the pentose phosphate pathway of glucose metabolism caused by hereditary deficiency of its key regulatory enzyme glucose-6-phosphate dehydrogenase (G6PD) has consequences that may worsen or attenuate the course of diabetic complications. Decreased availability of NADPH can predispose to oxidative stress and endothelial dysfunction, but can also limit the activity of the polyol pathway and cholesterol synthesis. Reduced availability of pentose phosphates for nucleic acid synthesis could impair cell proliferation. We sought to learn in which direction G6PD deficiency affects diabetic retinopathy. METHODS We enrolled patients who were G6PD-deficient or -sufficient with type 1 diabetes of duration 15 years or longer for whom HbA(1c) records were available for at least the previous 3 years. Renal failure and smoking were exclusion criteria. For each participant seven standard field colour photographs were obtained of each eye, and retinopathy was graded in a masked fashion. RESULTS The clinical characteristics of the 19 G6PD-deficient patients studied (age 42 ± 9 years, diabetes duration 24 ± 6 years, average HbA(1c) over 3 years 6.7 ± 0.8%) were similar to those of the 35 G6PD-sufficient patients. Almost 90% of patients in both groups had retinopathy; however, proliferative retinopathy was noted solely among G6PD-deficient patients (28%, p = 0.0036 vs G6PD-sufficient). The G6PD-deficient patients also showed a trend for increased frequency of microalbuminuria. CONCLUSIONS/INTERPRETATION The data suggest that G6PD deficiency accelerates the microvascular complications of diabetes, and that among the consequences of G6PD deficiency those that can enhance the damage caused by diabetes outweigh those that could be protective.
Collapse
Affiliation(s)
- G Cappai
- Ophthalmology Clinic, Ospedale San Giovanni di Dio, Cagliari, Italy
| | | | | | | | | |
Collapse
|
84
|
Bongard RD, Krenz GS, Gastonguay AJ, Williams CL, Lindemer BJ, Merker MP. Characterization of the threshold for NAD(P)H:quinone oxidoreductase activity in intact sulforaphane-treated pulmonary arterial endothelial cells. Free Radic Biol Med 2011; 50:953-62. [PMID: 21238579 PMCID: PMC3851029 DOI: 10.1016/j.freeradbiomed.2011.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 01/03/2011] [Accepted: 01/06/2011] [Indexed: 12/22/2022]
Abstract
Treatment of bovine pulmonary arterial endothelial cells in culture with the phase II enzyme inducer sulforaphane (5μM, 24h; sulf-treated) increased cell-lysate NAD(P)H:quinone oxidoreductase (NQO1) activity by 5.7 ± 0.6 (mean ± SEM)-fold, but intact-cell NQO1 activity by only 2.8 ± 0.1-fold compared to control cells. To evaluate the hypothesis that the threshold for sulforaphane-induced intact-cell NQO1 activity reflects a limitation in the capacity to supply NADPH at a sufficient rate to drive all the induced NQO1 to its maximum activity, total KOH-extractable pyridine nucleotides were measured in cells treated with duroquinone to stimulate maximal NQO1 activity. NQO1 activation increased NADP(+) in control and sulf-treated cells, with the effect more pronounced in the sulf-treated cells, in which the NADPH was also decreased. Glucose-6-phosphate dehydrogenase (G-6-PDH) inhibition partially blocked NQO1 activity in control and sulf-treated cells, but G-6-PDH overexpression via transient transfection with the human cDNA alleviated neither the restriction on intact sulf-treated cell NQO1 activity nor the impact on the NADPH/NADP(+) ratios. Intracellular ATP levels were not affected by NQO1 activation in control or sulf-treated cells. An increased dependence on extracellular glucose and a rightward shift in the K(m) for extracellular glucose were observed in NQO1-stimulated sulf-treated vs control cells. The data suggest that glucose transport in the sulf-treated cells may be insufficient to support the increased metabolic demand for pentose phosphate pathway-generated NADPH as an explanation for the NQO1 threshold.
Collapse
Affiliation(s)
- Robert D Bongard
- Department of Pulmonary Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
85
|
Gupte RS, Ata H, Rawat D, Abe M, Taylor MS, Ochi R, Gupte SA. Glucose-6-phosphate dehydrogenase is a regulator of vascular smooth muscle contraction. Antioxid Redox Signal 2011; 14:543-58. [PMID: 20649491 PMCID: PMC3029003 DOI: 10.1089/ars.2010.3207] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme in the pentose phosphate pathway and a major source of nicotinamide adenine dinucleotide phosphate reduced (NADPH), which regulates numerous enzymatic (including glutathione reductase and NADPH oxidase that, respectively, generates reduced glutathione and reactive oxygen species) reactions involved in various cellular actions, yet its physiological function is seldom investigated. We, however, recently showed that inhibiting G6PD causes precontracted coronary artery (CA) to relax in an endothelium-derived relaxing factor- and second messenger-independent manner. Here we assessed the role of G6PD in regulating CA contractility. Treating bovine CAs for 20 min with potassium chloride (KCl; 30 mM), amphotericin B (50 μM), or U46619 (100 nM) significantly (p < 0.05) increased both G6PD activity and glucose flux through the pentose phosphate pathway. The effect was Ca(2+) independent, and there was a corresponding increase in protein kinase C (PKC) activity. Activation of G6PD by KCl was blocked by the PKCδ inhibitor rottlerin (10 μM) or by knocking down PKCδ expression using siRNA. Phorbol 12, 13-dibutyrate (10 μM), a PKC activator, significantly increased G6PD phosphorylation and activity, whereas single (S210A, T266A) and double (S210A/T266A) mutations at sites flanking the G6PD active site significantly inhibited phosphorylation, shifted the isoelectric point, and reduced enzyme activity. Knocking down G6PD decreased NADPH and reactive oxygen species generation, and reduced KCl-evoked increases in [Ca(2+)](i) and myosin light chain phosphorylation, thereby reducing CA contractility. Similarly, aortas from G6PD-deficient mice developed less KCl/phorbol 12, 13-dibutyrate-evoked force than those from their wild-type littermates. Conversely, overexpression of G6PD augmented KCl-evoked increases in [Ca(2+)](i), thereby augmenting CA contraction. Our findings demonstrate that G6PD activity and NADPH is increased in activated CA in a PKCδ-dependent manner and that G6PD modulates Ca(2+) entry and CA contractions evoked by membrane depolarization.
Collapse
Affiliation(s)
- Rakhee S Gupte
- Department of Biochemistry, University of South Alabama, College of Medicine, Mobile, Alabama 36688, USA
| | | | | | | | | | | | | |
Collapse
|
86
|
Reciprocal regulation of 11β-hydroxysteroid dehydrogenase 1 and glucocorticoid receptor expression by dexamethasone inhibits human coronary artery smooth muscle cell proliferation in vitro. Mol Cell Biochem 2010; 346:69-79. [PMID: 20922465 DOI: 10.1007/s11010-010-0592-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Accepted: 09/18/2010] [Indexed: 10/19/2022]
Abstract
The actions of glucocorticoids are mediated, in part, by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), which amplifies their effects at the pre-receptor level by converting cortisone to cortisol. Glucocorticoids, such as dexamethasone, inhibit vascular smooth muscle cell proliferation; however, the role of 11β-HSD1 in this response remains unknown. Accordingly, we treated human coronary artery smooth muscle cells (HCSMC) with dexamethasone (10(-9)-10(-6) mol/l) and found that after 72 h dexamethasone increased 11β-HSD1 expression (14.16 ± 1.6-fold, P < 0.001) and activity (6.21 ± 1.2-fold, P < 0.001) in a dose- and time-dependent manner, which was dependent upon glucocorticoid receptor (GR) activation and C/EBPβ and C/EBPδ signaling. As glucocorticoids are known to negatively regulate GR expression, we examined the effect of decreasing 11β-HSD1 expression on GR expression. In HCSMC transfected with 11β-HSD1 siRNA, GR expression was increased; this effect was associated with protein kinase A activation and CREB phosphorylation. To examine the role of 11β-HSD1 in HCSMC proliferation, we decreased 11β-HSD1 expression and stimulated cells with platelet-derived growth factor (PDGF) (10 ng/ml). Decreased 11β-HSD1 expression was associated with increased cell proliferation in the absence of PDGF compared to scrambled control-transfected cells (236.10 ± 13.11%, n = 4, P < 0.001) and this effect was augmented by PDGF. Furthermore, the inhibitory effect of dexamethasone on cellular proliferation was abrogated in 11β-HSD1 siRNA-transfected HCSMC. Downregulation of 11β-HSD1 was associated with decreased p27(kip1) expression and increased phosphorylated retinoblastoma protein, consistent with a proliferative response. These findings suggest that 11β-HSD1 plays a role in the effects of glucocorticoids on vascular smooth muscle cell phenotype.
Collapse
|
87
|
Abstract
Dr. Joseph Loscalzo (M.D., 1978; Ph.D., 1977) is recognized here as a Redox Pioneer because he has published two articles in the field of antioxidant/redox biology that have been cited more than 1,000 times and 22 articles that have been cited more than 100 times. Dr. Loscalzo is known for his seminal contributions to our understanding of the vascular biology of nitric oxide. His initial discovery that the antiplatelet effects of organic nitrates are potentiated by thiols through a mechanism that involved metabolism to S-nitrosothiols was followed by the demonstration that S-nitrosothiols are formed endogenously through S-transnitrosation, stabilize nitric oxide, and facilitate the transport and transfer of nitric oxide between and within cells of the vessel wall. These properties led to the development of S-nitrosothiol-containing pharmacotherapies to treat disease states characterized by nitric oxide deficiency. Dr. Loscalzo's other scientific contributions include identifying the vascular functional consequences of genetic deficiencies of antioxidant enzymes that decrease nitric oxide bioavailability, collectively termed the "oxidative enzymopathies," and demonstrating the role of mitochondria in modulating the disulfide subproteome, and in redox signaling in hypoxia. He has received numerous awards and honors for his scientific contributions, including election to the Institute of Medicine of the National Academy of Sciences.
Collapse
Affiliation(s)
- Jane A Leopold
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
88
|
Abstract
According to a "canonical" view, reactive oxygen species (ROS) positively contribute, in different ways, to carcinogenesis and to malignant progression of tumor cells: they drive genomic damage and genetic instability, transduce, as signaling intermediates, mitogenic and survival inputs by growth factor receptors and adhesion molecules, promote cell motility and shape the tumor microenvironment by inducing inflammation/repair and angiogenesis. Chemopreventive and tumor-inhibitory effects of endogenous, diet-derived or supplemented antioxidants largely support this notion. However, emerging lines of evidence indicates that tumor cells also need to defend themselves from oxidative damage in order to survive and successfully spread at distance. This "heresy" has recently received important impulse from studies on the role of antioxidant capacity in cancer stem cells self-renewal and resistance to therapy; additionally, the transforming activity of some oncogenes has been unexpectedly linked to their capacity to maintain elevated intracellular levels of reduced glutathione (GSH), the principal redox buffer. These studies underline the importance of cellular antioxidant capacity in metastasis, as the result of a complex cell program involving enhanced motility and a profound change in energy metabolism. The glycolytic switch (Warburg effect) observed in malignant tissues is triggered by mitochondrial oxidative damage and/or activation of redox-sensitive transcription factors, and results in an increase of cell resistance to oxidants. On the other hand, cytoskeleton rearrangement underlying cell motile and tumor-aggressive behavior use ROS as intermediates and are therefore facilitated by oxidative stress. Along this line of speculation, we suggest that metastasis represents an integrated strategy for cancer cells to avoid oxidative damage and escape excess ROS in the primary tumor site, explaning why redox signaling pathways are often up-regulated in malignancy and metastasis.
Collapse
Affiliation(s)
- Giovambattista Pani
- Institute of General Pathology, Catholic University Medical School, Rome, Italy.
| | | | | |
Collapse
|
89
|
Benfotiamine improves functional recovery of the infarcted heart via activation of pro-survival G6PD/Akt signaling pathway and modulation of neurohormonal response. J Mol Cell Cardiol 2010; 49:625-38. [PMID: 20542491 DOI: 10.1016/j.yjmcc.2010.05.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 05/19/2010] [Accepted: 05/31/2010] [Indexed: 12/29/2022]
Abstract
Benfotiamine (BFT) is a transketolase activator that directs glucose to the pentose phosphate pathway. The present study investigated whether BFT improves the recovery after myocardial infarction (MI) and explored underlying mechanisms of protection. Non-diabetic and streptozotocin-induced type 1 diabetic mice were supplemented with BFT (70 mg/kg/day in drinking water) for 4 weeks and then subjected to MI or sham operation. Cardiac function was monitored by echocardiography. At two weeks post-MI, intra-ventricular pressure was measured by Millar tip-catheter and hearts were collected for biochemical, immunohistochemical and expressional analyses. No treatment effect was observed in sham-operated mice. Post-MI mortality was higher in diabetic mice and hemodynamic studies confirmed the worsening effect of diabetes on functional recovery. Furthermore, diabetic mice demonstrated increased cardiomyocyte apoptosis, reduced reparative angiogenesis, larger scars, enhanced oxidative stress, and blunted activation of the pro-survival VEGF receptor-2/Akt/Pim-1 signaling pathway. BFT improved post-MI survival, functional recovery and neovascularization and reduced cardiomyocyte apoptosis and neurohormonal activation in diabetic as well as in non-diabetic mice. In addition, BFT stimulated the activity of pentose phosphate pathway enzymes, leading to reduction of oxidative stress, phosphorylation/activation of VEGF receptor-2 and Akt and increased Pim-1, pBad and Bcl-2 levels. These effects were contrasted on silencing glucose-6-phosphate dehydrogenase, the key enzyme in pentose phosphate pathway, or inhibiting Akt. BFT benefits post-MI recovery through stimulation of pro-survival mechanisms and containment of neurohormonal response. These results may have implications for the treatment of myocardial ischemia.
Collapse
|
90
|
Skvortsova I, Skvortsov S, Raju U, Stasyk T, Riesterer O, Schottdorf EM, Popper BA, Schiestl B, Eichberger P, Debbage P, Neher A, Bonn GK, Huber LA, Milas L, Lukas P. Epithelial-to-mesenchymal transition and c-myc expression are the determinants of cetuximab-induced enhancement of squamous cell carcinoma radioresponse. Radiother Oncol 2010; 96:108-15. [PMID: 20451273 DOI: 10.1016/j.radonc.2010.04.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 03/24/2010] [Accepted: 04/14/2010] [Indexed: 11/16/2022]
Abstract
PURPOSE Radiation therapy cures malignant tumors of the head and neck region more effectively when it is combined with application of the anti-EGFR monoclonal antibody cetuximab. Despite the successes achieved, we still do not know how to select patients who will respond to this combination of anti-EGFR monoclonal antibody and radiation. This study was conducted to elucidate possible mechanisms which cause the combined treatment with cetuximab and irradiation to fail in some cases of squamous cell carcinomas. METHODS AND MATERIALS Mice bearing FaDu and A431 squamous cell carcinoma xenograft tumors were treated with cetuximab (total dose 3 mg, intraperitoneally), irradiation (10 Gy) or their combination at the same doses. Treatment was applied when tumors reached 8mm in size. To collect samples for further protein analysis (two-dimensional differential gel electrophoresis (2-D DIGE), mass spectrometry MALDI-TOF/TOF, Western blot analysis, and ELISA), mice from each group were sacrificed on the 8th day after the first injection of cetuximab. Other mice were subjected to tumor growth delay assay. RESULTS In FaDu xenografts, treatment with cetuximab alone was nearly as effective as cetuximab combined with ionizing radiation, whereas A431 tumors responded to the combined treatment with significantly enhanced delay in tumor growth. Tumors extracted from the untreated FaDu and A431 xenografts were analysed for protein expression, and 34 proteins that were differently expressed in the two tumor types were identified. The majority of these proteins are closely related to intratumoral angiogenesis, cell adhesion, motility, differentiation, epithelial-to-mesenchymal transition (EMT), c-myc signaling and DNA repair. CONCLUSIONS The failure of cetuximab to enhance radiation response in FaDu xenografts was associated with the initiation of the program of EMT and with c-myc up-regulation in the carcinoma cells. For this reason, c-myc and EMT-related proteins (E-cadherin, vimentin) may be considered as potential biomarkers to predict squamous cell carcinoma response after treatment with cetuximab in combination with radiation.
Collapse
Affiliation(s)
- Ira Skvortsova
- Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Abstract
Cellular respiration in an oxygen-rich environment leads to the generation of reactive oxygen species. These partially reduced forms of molecular oxygen can readily react with biological molecules, often modifying their normal biological function. Antioxidant enzyme mechanisms have evolved to eliminate reactive oxygen species and minimize the oxidant stress caused by their reactivity. Inherited and acquired deficiencies of key antioxidant enzymes lead to a dysregulated redox environment, which can promote pathobiology; when this redox dysfunction occurs in the blood vessel, vascular disease ensues. In this article, we consider three distinct antioxidant enzyme deficiencies - glucose-6-phosphate dehydrogenase, glutathione peroxidase-1 and glutathione peroxidase-3 - and their consequences for vascular disease.
Collapse
Affiliation(s)
- Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA Tel.: +1 617 732 6340
| |
Collapse
|
92
|
Zhang Z, Liew CW, Handy DE, Zhang Y, Leopold JA, Hu J, Guo L, Kulkarni RN, Loscalzo J, Stanton RC. High glucose inhibits glucose-6-phosphate dehydrogenase, leading to increased oxidative stress and beta-cell apoptosis. FASEB J 2009; 24:1497-505. [PMID: 20032314 DOI: 10.1096/fj.09-136572] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patients with type 2 diabetes lose beta cells, but the underlying mechanisms are incompletely understood. Glucose-6-phosphate dehydrogenase (G6PD) is the principal source of the major intracellular reductant, NADPH, which is required by many enzymes, including enzymes of the antioxidant pathway. Previous work from our laboratory has shown that high glucose impairs G6PD activity in endothelial and kidney cells, which leads to decreased cell survival. Pancreatic beta cells are highly sensitive to increased ROS. This study aimed to determine whether G6PD and NADPH play central roles in beta-cell survival. Human and mouse islets, MIN6 cell line, and G6PD deficient mice were studied. High glucose inhibited G6PD expression and activity. Inhibition of G6PD with siRNA led to increased ROS and apoptosis, decreased proliferation, and impaired insulin secretion. High glucose decreased insulin secretion, which was improved by overexpressing G6PD. G6PD-deficient mice had smaller islets and impaired glucose tolerance compared with control mice, which suggests that G6PD deficiency per se leads to beta-cell dysfunction and death. G6PD plays an important role in beta-cell function and survival. High-glucose-mediated decrease in G6PD activity may provide a mechanistic explanation for the gradual loss of beta cells in patients with diabetes.
Collapse
Affiliation(s)
- Zhaoyun Zhang
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Leopold JA, Loscalzo J. Oxidative risk for atherothrombotic cardiovascular disease. Free Radic Biol Med 2009; 47:1673-706. [PMID: 19751821 PMCID: PMC2797369 DOI: 10.1016/j.freeradbiomed.2009.09.009] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 08/31/2009] [Accepted: 09/06/2009] [Indexed: 02/07/2023]
Abstract
In the vasculature, reactive oxidant species, including reactive oxygen, nitrogen, or halogenating species, and thiyl, tyrosyl, or protein radicals may oxidatively modify lipids and proteins with deleterious consequences for vascular function. These biologically active free radical and nonradical species may be produced by increased activation of oxidant-generating sources and/or decreased cellular antioxidant capacity. Once formed, these species may engage in reactions to yield more potent oxidants that promote transition of the homeostatic vascular phenotype to a pathobiological state that is permissive for atherothrombogenesis. This dysfunctional vasculature is characterized by lipid peroxidation and aberrant lipid deposition, inflammation, immune cell activation, platelet activation, thrombus formation, and disturbed hemodynamic flow. Each of these pathobiological states is associated with an increase in the vascular burden of free radical species-derived oxidation products and, thereby, implicates increased oxidant stress in the pathogenesis of atherothrombotic vascular disease.
Collapse
Affiliation(s)
- Jane A Leopold
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
94
|
Xu Y, Zhang Z, Hu J, Stillman IE, Leopold JA, Handy DE, Loscalzo J, Stanton RC. Glucose-6-phosphate dehydrogenase-deficient mice have increased renal oxidative stress and increased albuminuria. FASEB J 2009; 24:609-16. [PMID: 19805580 DOI: 10.1096/fj.09-135731] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme of the pentose phosphate pathway and the principal source of NADPH, a major cellular reductant, and is central to cell survival. Our previous work showed that diabetes and increased aldosterone are acquired forms of G6PD deficiency, leading to decreased G6PD activity and NADPH levels and damage to kidney tissue and endothelial cells. In this study, G6PD-deficient mice were studied to test the hypothesis that decreased G6PD activity per se can cause changes similar to those seen in the acquired conditions of G6PD deficiency. Results show that as compared with control mice, G6PD-deficient mice had increased oxidative stress, as manifested by decreased NADPH levels and decreased GSH levels, and increased markers of lipid peroxidation. G6PD-deficient mice had increased protein kinase C activity, increased nuclear factor-kappaB activity, and increased urinary albumin levels, all of which is similar to changes seen in diabetic mice. Changes persisted as the mice aged, as old G6PD-deficient mice (17-20 mo) had higher urine albumin levels and also had evidence for increased apoptosis in the renal cortex. These results show that decreased G6PD activity per se is sufficient to cause changes similar to those seen in diabetic mice.
Collapse
Affiliation(s)
- Yizhen Xu
- Renal Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Bonnet S, Paulin R, Sutendra G, Dromparis P, Roy M, Watson KO, Nagendran J, Haromy A, Dyck JRB, Michelakis ED. Dehydroepiandrosterone reverses systemic vascular remodeling through the inhibition of the Akt/GSK3-{beta}/NFAT axis. Circulation 2009; 120:1231-40. [PMID: 19752325 DOI: 10.1161/circulationaha.109.848911] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The remodeled vessel wall in many vascular diseases such as restenosis after injury is characterized by proliferative and apoptosis-resistant vascular smooth muscle cells. There is evidence that proproliferative and antiapoptotic states are characterized by a metabolic (glycolytic phenotype and hyperpolarized mitochondria) and electric (downregulation and inhibition of plasmalemmal K(+) channels) remodeling that involves activation of the Akt pathway. Dehydroepiandrosterone (DHEA) is a naturally occurring and clinically used steroid known to inhibit the Akt axis in cancer. We hypothesized that DHEA will prevent and reverse the remodeling that follows vascular injury. METHODS AND RESULTS We used cultured human carotid vascular smooth muscle cell and saphenous vein grafts in tissue culture, stimulated by platelet-derived growth factor to induce proliferation in vitro and the rat carotid injury model in vivo. DHEA decreased proliferation and increased vascular smooth muscle cell apoptosis in vitro and in vivo, reducing vascular remodeling while sparing healthy tissues after oral intake. Using pharmacological (agonists and antagonists of Akt and its downstream target glycogen-synthase-kinase-3beta [GSK-3beta]) and molecular (forced expression of constitutively active Akt1) approaches, we showed that the effects of DHEA were mediated by inhibition of Akt and subsequent activation of GSK-3beta, leading to mitochondrial depolarization, increased reactive oxygen species, activation of redox-sensitive plasmalemmal voltage-gated K(+) channels, and decreased [Ca(2+)](i). These functional changes were accompanied by sustained molecular effects toward the same direction; by decreasing [Ca(2+)](i) and inhibiting GSK-3beta, DHEA inhibited the nuclear factor of activated T cells transcription factor, thus increasing expression of Kv channels (Kv1.5) and contributing to sustained mitochondrial depolarization. These results were independent of any steroid-related effects because they were not altered by androgen and estrogen inhibitors but involved a membrane G protein-coupled receptor. CONCLUSIONS We suggest that the orally available DHEA might be an attractive candidate for the treatment of systemic vascular remodeling, including restenosis, and we propose a novel mechanism of action for this important hormone and drug.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Centre de Recherche de L'Hôtel-Dieu de Québec, 9 Rue McMahon, Québec, Qc, G1R 2J6, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Pan S, World CJ, Kovacs CJ, Berk BC. Glucose 6-Phosphate Dehydrogenase Is Regulated Through c-Src–Mediated Tyrosine Phosphorylation in Endothelial Cells. Arterioscler Thromb Vasc Biol 2009; 29:895-901. [DOI: 10.1161/atvbaha.109.184812] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Shi Pan
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, NY
| | - Cameron J. World
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, NY
| | - Christopher J. Kovacs
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, NY
| | - Bradford C. Berk
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
97
|
Vizán P, Sánchez-Tena S, Alcarraz-Vizán G, Soler M, Messeguer R, Pujol MD, Lee WNP, Cascante M. Characterization of the metabolic changes underlying growth factor angiogenic activation: identification of new potential therapeutic targets. Carcinogenesis 2009; 30:946-52. [PMID: 19369582 DOI: 10.1093/carcin/bgp083] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is a fundamental process to normal and abnormal tissue growth and repair, which consists of recruiting endothelial cells toward an angiogenic stimulus. The cells subsequently proliferate and differentiate to form endothelial tubes and capillary-like structures. Little is known about the metabolic adaptation of endothelial cells through such a transformation. We studied the metabolic changes of endothelial cell activation by growth factors using human umbilical vein endothelial cells (HUVECs), [1,2-(13)C(2)]-glucose and mass isotopomer distribution analysis. The metabolism of [1,2-(13)C(2)]-glucose by HUVEC allows us to trace many of the main glucose metabolic pathways, including glycogen synthesis, the pentose cycle and the glycolytic pathways. So we established that these pathways were crucial to endothelial cell proliferation under vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) stimulation. A specific VEGF receptor-2 inhibitor demonstrated the importance of glycogen metabolism and pentose cycle pathway. Furthermore, we showed that glycogen was depleted in a low glucose medium, but conserved under hypoxic conditions. Finally, we demonstrated that direct inhibition of key enzymes to glycogen metabolism and pentose phosphate pathways reduced HUVEC viability and migration. In this regard, inhibitors of these pathways have been shown to be effective antitumoral agents. To sum up, our data suggest that the inhibition of metabolic pathways offers a novel and powerful therapeutic approach, which simultaneously inhibits tumor cell proliferation and tumor-induced angiogenesis.
Collapse
Affiliation(s)
- Pedro Vizán
- Department of Biochemistry and Molecular Biology, University of Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Handy DE, Lubos E, Yang Y, Galbraith JD, Kelly N, Zhang YY, Leopold JA, Loscalzo J. Glutathione peroxidase-1 regulates mitochondrial function to modulate redox-dependent cellular responses. J Biol Chem 2009; 284:11913-21. [PMID: 19254950 DOI: 10.1074/jbc.m900392200] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Glutathione peroxidase-1 (GPx-1) is a selenocysteine-containing enzyme that plays a major role in the reductive detoxification of peroxides in cells. In permanently transfected cells with approximate 2-fold overexpression of GPx-1, we found that intracellular accumulation of oxidants in response to exogenous hydrogen peroxide was diminished, as was epidermal growth factor receptor (EGFR)-mediated Akt activation in response to hydrogen peroxide or EGF stimulation. Knockdown of GPx-1 augmented EGFR-mediated Akt activation, whereas overexpression of catalase decreased Akt activation, suggesting that EGFR signaling is regulated by redox mechanisms. To determine whether mitochondrial oxidants played a role in these processes, cells were pretreated with a mitochondrial uncoupler prior to EGF stimulation. Inhibition of mitochondrial function attenuated EGF-mediated activation of Akt in control cells but had no additional effect in GPx-1-overexpressing cells, suggesting that GPx-1 overexpression decreased EGFR signaling by decreasing mitochondrial oxidants. Consistent with this finding, GPx-1 overexpression decreased global protein disulfide bond formation, which is dependent on mitochondrially produced oxidants. GPx-1 overexpression, in permanently transfected or adenovirus-treated cells, also caused overall mitochondrial dysfunction with a decrease in mitochondrial potential and a decrease in ATP production. GPx-1 overexpression also decreased EGF- and serum-mediated [(3)H]thymidine incorporation, indicating that alterations in GPx-1 can attenuate cell proliferation. Taken together, these data suggest that GPx-1 can modulate redox-dependent cellular responses by regulating mitochondrial function.
Collapse
Affiliation(s)
- Diane E Handy
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
99
|
The behavior of endothelial cells on polyurethane nanocomposites and the associated signaling pathways. Biomaterials 2009; 30:1502-11. [PMID: 19118895 DOI: 10.1016/j.biomaterials.2008.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 12/01/2008] [Indexed: 11/23/2022]
Abstract
A series of nanocomposites from polyurethane (PU) incorporated with various low concentrations (17.4-174 ppm) of gold nanoparticles (approximately 5 nm) (denoted "PU-Au") were used as a model system to study the mechanisms that influenced endothelial cell (EC) migration on biomaterial surfaces. The migration rate of ECs on the PU-Au nanocomposites was determined by a real-time image system. It was found that ECs had the highest migration rate on the nanocomposite containing 43.5 ppm of gold ("PU-Au 43.5 ppm"). The high EC migration rate was associated with increased levels of endothelial nitric oxide synthase (eNOS) and phosphorylated-Akt (p-Akt) expressed by ECs cultured on PU-Au. The inductions of both eNOS and p-Akt on PU-Au were abolished by the addition of LY294002 (PI3K inhibitor), suggesting that these cellular events may be regulated through the PI3K signaling pathway. Using a biotinylated VEGF-165 that recognizes VEGF receptors and by FACS analysis, slightly higher expression of VEGF receptors for ECs on PU-Au was also demonstrated. Phalloidin staining showed that actin appeared as a circumferential band surrounding each cell on tissue culture polystyrene, whereas on PU-Au, especially on PU-Au 43.5 ppm, the cells had their margin spread out and extend processes with stress fibers in the protruding lamellipodia. Moreover, the higher EC migration rate on PU-Au 43.5 ppm was suppressed by LY294002. The higher protein expression of focal adhesion kinase (FAK) on PU-Au 43.5 ppm was observed in FAK-GFP transfected ECs. It was concluded that PU-Au nanocomposites activated FAK and the PI3K/Akt signaling pathway in ECs, leading to proliferation and migration of ECs on these surfaces.
Collapse
|
100
|
Ravi Kumar GVPPS, Srinivas G, Sharma A, Suryanarayana VVS, Ravi Kumar P, Bhattacharya TK, Mitra A. Sequencing, characterization and genetic variation of the Bos indicus glucose-6-phosphate-dehydrogenase gene. DNA SEQUENCE : THE JOURNAL OF DNA SEQUENCING AND MAPPING 2008; 19:37-43. [PMID: 18300160 DOI: 10.1080/10425170701283977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The coding sequence of the bovine (Bos indicus) Glucose-6-phosphate-dehydrogenase (G6PD) gene was amplified by Reverse Transcriptase-PCR (RT-PCR), cloned, sequenced and characterized. The deduced amino acid sequence clustered the bovine G6PD sequence with the other mammalian G6PD proteins into a monophyletic group. The bovids (B. indicus and B. taurus) clustered clearly from the rodent (rat, mouse and hamster) subcluster and from humans. The multiple sequence alignment of the bovine G6PD with the mammalian species clearly revealed conservation of the substrate, coenzyme, catalytic and the dimer binding sites with the solved X-ray crystallographic structure of Homo sapiens. Also, four fragments of bovine (Bos indicus) G6PD gene viz. 118, 319, 683 and 408 bp were amplified and sequenced for the first time. A G/A and G/C single nucleotide polymorphisms in intron-9 and exon-10 were detected on PCR-RFLP of the 319 bp amplicon with Hae III and Pst I, respectively. This work is the first study on Bos indicus G6PD gene at the nucleotide level.
Collapse
Affiliation(s)
- G V P P S Ravi Kumar
- Molecular Cytogenetics Lab, Department of Animal Breeding and Genetics, GADVASU, PAU Campus, Ludhiana 141004, India.
| | | | | | | | | | | | | |
Collapse
|