1
|
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F, Lamas S. Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med 2024; 222:85-105. [PMID: 38838921 DOI: 10.1016/j.freeradbiomed.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.
Collapse
Affiliation(s)
- Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Fernando Rodríguez-Pascual
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
2
|
Zhou LY, Liu K, Yin WJ, Xie YL, Wang JL, Zuo SR, Tang ZY, Wu YF, Zuo XC. Arginase2 mediates contrast-induced acute kidney injury via facilitating nitrosative stress in tubular cells. Redox Biol 2023; 67:102929. [PMID: 37856999 PMCID: PMC10587771 DOI: 10.1016/j.redox.2023.102929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023] Open
Abstract
Contrast-induced acute kidney injury(CI-AKI) is the third cause of AKI. Although tubular injury has been regarded as an important pathophysiology of CI-AKI, the underlying mechanism remains elusive. Here, we found arginase2(ARG2) accumulated in the tubules of CI-AKI mice, and was upregulated in iohexol treated kidney tubular cells and in blood samples of CI-AKI mice and patients, accompanied by increased nitrosative stress and apoptosis. However, all of the above were reversed in ARG2 knockout mice, as evidenced by the ameliorated kidney dysfunction and the tubular injury, and decreased nitrosative stress and apoptosis. Mechanistically, HO-1 upregulation could alleviate iohexol or ARG2 overexpression mediated nitrosative stress. Silencing and overexpressing ARG2 was able to upregulate and downregulate HO-1 expression, respectively, while HO-1 siRNA had no effect on ARG2 expression, indicating that ARG2 might inhibit HO-1 expression at the transcriptional level, which facilitated nitrosative stress during CI-AKI. Additionally, CREB1, a transcription factor, bound to the promoter region of ARG2 and stimulated its transcription. Similar findings were yielded in cisplatin- or vancomycin-induced AKI models. Taken together, ARG2 is a crucial target of CI-AKI, and activating CREB1/ARG2/HO-1 axis can mediate tubular injury by promoting nitrosative stress, highlighting potential therapeutic strategy for treating CI-AKI.
Collapse
Affiliation(s)
- Ling-Yun Zhou
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Kun Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Jun Yin
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yue-Liang Xie
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiang-Lin Wang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shan-Ru Zuo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Yao Tang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi-Feng Wu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Cong Zuo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China; Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Guo T, Hu S, Xu W, Zhou J, Chen F, Gao T, Qu W, Chen F, Lv Z, Lu L. Elevated expression of histone deacetylase HDAC8 suppresses arginine-proline metabolism in necrotizing enterocolitis. iScience 2023; 26:106882. [PMID: 37260741 PMCID: PMC10227426 DOI: 10.1016/j.isci.2023.106882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/07/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
Epigenetic alterations are especially important in necrotizing enterocolitis (NEC). Here, we reported that histone deacetylase 8 (HDAC8) plays a previously unknown role in modulating arginine metabolism via acetylation of histone 3 lysine 9 (acetyl-H3K9) regulation during the pathogenesis of NEC. We found that HDAC8 was upregulated in humans and mice intestinal samples with NEC, while selective inhibition of HDAC8 expression ameliorated NEC. HDAC8 regulates enzymes involved in the metabolic conversion of proline to arginine (PRODH, PRODH2, OAT, and OTC) and arginine to ornithine (ARG1). The results showed that H3K9ac signal in the PRODH/PRODH2 promoter region was mediated by HDAC8. Additionally, the decreased concentration of butyric acid was strongly correlated with elevated HDAC8 levels and circulating arginine, which may result from an unbalanced Firmicutes/Bacteroidetes ratio. These results reveal previously underappreciated roles of microbial metabolites and HDAC8 to coordinate the arginine metabolism during NEC development.
Collapse
Affiliation(s)
- Ting Guo
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Shaohua Hu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Weijue Xu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Jin Zhou
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Feng Chen
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Tingting Gao
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Wenqian Qu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Faling Chen
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Li Lu
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200040, China
| |
Collapse
|
4
|
Douglass MS, Kaplowitz MR, Zhang Y, Fike CD. Impact of l-citrulline on nitric oxide signaling and arginase activity in hypoxic human pulmonary artery endothelial cells. Pulm Circ 2023; 13:e12221. [PMID: 37063746 PMCID: PMC10091859 DOI: 10.1002/pul2.12221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
Impaired nitric oxide (NO) signaling contributes to the development of pulmonary hypertension (PH). The l-arginine precursor, l-citrulline, improves NO signaling and has therapeutic potential in PH. However, there is evidence that l-citrulline might increase arginase activity, which in turn, has been shown to contribute to PH. Our major purpose was to determine if l-citrulline increases arginase activity in hypoxic human pulmonary artery endothelial cells (PAECs). In addition, to avoid potential adverse effects from high dose l-citrulline monotherapy, we evaluated whether the effect on NO signaling is greater using co-treatment with l-citrulline and another agent that improves NO signaling, folic acid, than either alone. Arginase activity was measured in human PAECs cultured under hypoxic conditions in the presence of l-citrulline (0-1 mM). NO production and endothelial nitric oxide synthase (eNOS) coupling, as assessed by eNOS dimer-to-monomer ratios, were measured in PAECs treated with l-citrulline and/or folic acid (0.2 μM). Arginase activity increased in hypoxic PAECs treated with 1 mM but not with either 0.05 or 0.1 mM l-citrulline. Co-treatment with folic acid and 0.1 mM l-citrulline increased NO production and eNOS dimer-to-monomer ratios more than treatment with either alone. The potential to increase arginase activity suggests that there might be plasma l-citrulline concentrations that should not be exceeded when using l-citrulline to treat PH. Rather than progressively increasing the dose of l-citrulline as a monotherapy, co-therapy with l-citrulline and folic acid merits consideration, due to the possibility of achieving efficacy at lower doses and minimizing side effects.
Collapse
Affiliation(s)
| | | | - Yongmei Zhang
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Candice D. Fike
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
5
|
Ma Y, Potenza DM, Ajalbert G, Brenna A, Zhu C, Ming XF, Yang Z. Paracrine Effects of Renal Proximal Tubular Epithelial Cells on Podocyte Injury under Hypoxic Conditions Are Mediated by Arginase-II and TGF-β1. Int J Mol Sci 2023; 24:ijms24043587. [PMID: 36835007 PMCID: PMC9966309 DOI: 10.3390/ijms24043587] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Hypoxia is an important risk for renal disease. The mitochondrial enzyme arginase-II (Arg-II) is expressed and/or induced by hypoxia in proximal tubular epithelial cells (PTECs) and in podocytes, leading to cellular damage. Because PTECs are vulnerable to hypoxia and located in proximity to podocytes, we examined the role of Arg-II in the crosstalk of PTECs under hypoxic conditions with podocytes. A human PTEC cell line (HK2) and a human podocyte cell line (AB8/13) were cultured. Arg-ii gene was ablated by CRISPR/Case9 in both cell types. HK2 cells were exposed to normoxia (21% O2) or hypoxia (1% O2) for 48 h. Conditioned medium (CM) was collected and transferred to the podocytes. Podocyte injuries were then analyzed. Hypoxic (not normoxic) HK2-CM caused cytoskeletal derangement, cell apoptosis, and increased Arg-II levels in differentiated podocytes. These effects were absent when arg-ii in HK2 was ablated. The detrimental effects of the hypoxic HK2-CM were prevented by TGF-β1 type-I receptor blocker SB431542. Indeed, TGF-β1 levels in hypoxic HK2-CM (but not arg-ii-/--HK2-CM) were increased. Furthermore, the detrimental effects of TGF-β1 on podocytes were prevented in arg-ii-/--podocytes. This study demonstrates crosstalk between PTECs and podocytes through the Arg-II-TGF-β1 cascade, which may contribute to hypoxia-induced podocyte damage.
Collapse
|
6
|
Ren Z, Potenza DM, Ma Y, Ajalbert G, Hoogewijs D, Ming XF, Yang Z. Role of Arginase-II in Podocyte Injury under Hypoxic Conditions. Biomolecules 2022; 12:biom12091213. [PMID: 36139052 PMCID: PMC9496188 DOI: 10.3390/biom12091213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia plays a crucial role in acute and chronic renal injury, which is attributable to renal tubular and glomerular cell damage. Some studies provide evidence that hypoxia-dependent upregulation of the mitochondrial enzyme arginase type-II (Arg-II) in tubular cells promotes renal tubular injury. It is, however, not known whether Arg-II is also expressed in glomerular cells, particularly podocytes under hypoxic conditions, contributing to hypoxia-induced podocyte injury. The effects of hypoxia on human podocyte cells (AB8/13) in cultures and on isolated kidneys from wild-type (wt) and arg-ii gene-deficient (arg-ii−/−) mice ex vivo, as well as on mice of the two genotypes in vivo, were investigated, respectively. We found that the Arg-II levels were enhanced in cultured podocytes in a time-dependent manner over 48 h, which was dependent on the stabilization of hypoxia-inducible factor 1α (HIF1α). Moreover, a hypoxia-induced derangement of cellular actin cytoskeletal fibers, a decrease in podocin, and an increase in mitochondrial ROS (mtROS) generation—as measured by MitoSOX—were inhibited by adenoviral-mediated arg-ii gene silencing. These effects of hypoxia on podocyte injury were mimicked by the HIFα stabilizing drug DMOG, which inhibits prolyl hydroxylases (PHD), the enzymes involved in HIFα degradation. The silencing of arg-ii prevented the detrimental effects of DMOG on podocytes. Furthermore, the inhibition of mtROS generation by rotenone—the inhibitor of respiration chain complex-I—recapitulated the protective effects of arg-ii silencing on podocytes under hypoxic conditions. Moreover, the ex vivo experiments with isolated kidney tissues and the in vivo experiments with mice exposed to hypoxic conditions showed increased Arg-II levels in podocytes and decreased podocyte markers regarding synaptopodin in wt mice but not in arg-ii−/− mice. While age-associated albuminuria was reduced in the arg-ii−/− mice, the hypoxia-induced increase in albuminuria was, however, not significantly affected in the arg-ii−/−. Our study demonstrates that Arg-II in podocytes promotes cell injury. Arg-ii ablation seems insufficient to protect mice in vivo against a hypoxia-induced increase in albuminuria, but it does reduce albuminuria in aging.
Collapse
Affiliation(s)
- Zhilong Ren
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Duilio Michele Potenza
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Yiqiong Ma
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guillaume Ajalbert
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - David Hoogewijs
- Integrative Oxygen Physiology, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Xiu-Fen Ming
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
- Correspondence: (X.-F.M.); (Z.Y.); Tel.: +41-26-300-85-93 (Z.Y.)
| | - Zhihong Yang
- Cardiovascular & Aging Research, Department of Endocrinology, Metabolism, Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
- Correspondence: (X.-F.M.); (Z.Y.); Tel.: +41-26-300-85-93 (Z.Y.)
| |
Collapse
|
7
|
Modulation of Arginase-2 mRNA Levels by ω-3 PUFAs and Aspirin in Asthmatic Human Lung Fibroblasts. J Lipids 2022; 2022:3062274. [PMID: 36061615 PMCID: PMC9436553 DOI: 10.1155/2022/3062274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/05/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Airway remodeling (AR) increases disease severity, and morbidity of asthmatic patients by contributing to irreversible airflow obstruction and progressive declines in lung function. Arginase isoenzymes and the downstream enzymes ornithine decarboxylase (ODC) and ornithine aminotransferase (OAT) have been implicated in the hyperplastic and fibrotic changes of AR, respectively. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) and resolvin metabolites have anti-AR effects, but whether they are mediated through the arginase pathway is unclear. Our study intended to determine the effects of the ω-3 PUFAs eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), resolvin D1 (RvD1), TH1 cytokines, acetylsalicylic acid (ASA), cAMP, and dexamethasone (DEX) on the expression of arginase isoenzymes arginase 1 (ARG1) and arginase 2 (ARG2), ODC, and OAT in human lung fibroblasts (HLF) from normal (NHLF) and diseased (DHLF) asthmatic donors using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR). Our data showed that EPA and EPA+DHA downregulated ARG2 mRNA 2-fold in both types of HLF. DHA, RvD1, and DEX did not alter mRNA levels for any of the genes studied. EPA lowered the ARG2 protein levels in DHLF, but did not affect those levels in NHLF. ASA upregulated ARG2 mRNA 5-fold and 7-fold in NHLF and DHLF, respectively, TH1 cytokines downregulated ARG2, ODC, and OAT mRNA in DHLF 10-fold, 2-fold, and 2.5-fold, respectively, and cAMP downregulated ARG2 mRNA 2-fold in DHLF. These results are the first to show a direct effect of ω-3 PUFAs on ARG2 mRNA levels and provide further evidence for a role of ω-3 PUFAs in AR.
Collapse
|
8
|
Reiterer M, Eakin A, Johnson RS, Branco CM. Hyperoxia Reprogrammes Microvascular Endothelial Cell Response to Hypoxia in an Organ-Specific Manner. Cells 2022; 11:cells11162469. [PMID: 36010546 PMCID: PMC9406746 DOI: 10.3390/cells11162469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Organ function relies on microvascular networks to maintain homeostatic equilibrium, which varies widely in different organs and during different physiological challenges. The endothelium role in this critical process can only be evaluated in physiologically relevant contexts. Comparing the responses to oxygen flux in primary murine microvascular EC (MVEC) obtained from brain and lung tissue reveals that supra-physiological oxygen tensions can compromise MVEC viability. Brain MVEC lose mitochondrial activity and undergo significant alterations in electron transport chain (ETC) composition when cultured under standard, non-physiological atmospheric oxygen levels. While glycolytic capacity of both lung and brain MVEC are unchanged by environmental oxygen, the ability to trigger a metabolic shift when oxygen levels drop is greatly compromised following exposure to hyperoxia. This is particularly striking in MVEC from the brain. This work demonstrates that the unique metabolism and function of organ-specific MVEC (1) can be reprogrammed by external oxygen, (2) that this reprogramming can compromise MVEC survival and, importantly, (3) that ex vivo modelling of endothelial function is significantly affected by culture conditions. It further demonstrates that physiological, metabolic and functional studies performed in non-physiological environments do not represent cell function in situ, and this has serious implications in the interpretation of cell-based pre-clinical models.
Collapse
Affiliation(s)
- Moritz Reiterer
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda Eakin
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
| | - Randall S. Johnson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Cristina M. Branco
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK
- Correspondence:
| |
Collapse
|
9
|
Liang X, Potenza DM, Brenna A, Ma Y, Ren Z, Cheng X, Ming XF, Yang Z. Hypoxia Induces Renal Epithelial Injury and Activates Fibrotic Signaling Through Up-Regulation of Arginase-II. Front Physiol 2021; 12:773719. [PMID: 34867480 PMCID: PMC8640467 DOI: 10.3389/fphys.2021.773719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
The ureohydrolase, type-II arginase (Arg-II), is a mitochondrial enzyme metabolizing L-arginine into urea and L-ornithine and is highly expressed in renal proximal tubular cells (PTC) and upregulated by renal ischemia. Recent studies reported contradictory results on the role of Arg-II in renal injury. The aim of our study is to investigate the function of Arg-II in renal epithelial cell damage under hypoxic conditions. Human renal epithelial cell line HK2 was cultured under hypoxic conditions for 12–48 h. Moreover, ex vivo experiments with isolated kidneys from wild-type (WT) and genetic Arg-II deficient mice (Arg-II–/–) were conducted under normoxic and hypoxic conditions. The results show that hypoxia upregulates Arg-II expression in HK2 cells, which is inhibited by silencing both hypoxia-inducible factors (HIFs) HIF1α and HIF2α. Treatment of the cells with dimethyloxaloylglycine (DMOG) to stabilize HIFα also enhances Arg-II. Interestingly, hypoxia or DMOG upregulates transforming growth factor β1 (TGFβ1) levels and collagens Iα1, which is prevented by Arg-II silencing, while TGFβ1-induced collagen Iα1 expression is not affected by Arg-II silencing. Inhibition of mitochondrial complex-I by rotenone abolishes hypoxia-induced reactive oxygen species (mtROS) and TGFβ1 elevation in the cells. Ex vivo experiments show elevated Arg-II and TGFβ1 expression and the injury marker NGAL in the WT mouse kidneys under hypoxic conditions, which is prevented in the Arg-II–/– mice. Taking together, the results demonstrate that hypoxia activates renal epithelial HIFs-Arg-II-mtROS-TGFβ1-cascade, participating in hypoxia-associated renal injury and fibrosis.
Collapse
Affiliation(s)
- Xiujie Liang
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Duilio Michele Potenza
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Andrea Brenna
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Yiqiong Ma
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Zhilong Ren
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Xin Cheng
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Xiu-Fen Ming
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Zhihong Yang
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
10
|
Li J, Fang Y, Wu D. Mechanical forces and metabolic changes cooperate to drive cellular memory and endothelial phenotypes. CURRENT TOPICS IN MEMBRANES 2021; 87:199-253. [PMID: 34696886 PMCID: PMC8639155 DOI: 10.1016/bs.ctm.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial cells line the innermost layer of arterial, venous, and lymphatic vascular tree and accordingly are subject to hemodynamic, stretch, and stiffness mechanical forces. Normally quiescent, endothelial cells have a hemodynamic set point and become "activated" in response to disturbed hemodynamics, which may signal impending nutrient or gas depletion. Endothelial cells in the majority of tissue beds are normally inactivated and maintain vessel barrier functions, are anti-inflammatory, anti-coagulant, and anti-thrombotic. However, under aberrant mechanical forces, endothelial signaling transforms in response, resulting cellular changes that herald pathological diseases. Endothelial cell metabolism is now recognized as the primary intermediate pathway that undergirds cellular transformation. In this review, we discuss the various mechanical forces endothelial cells sense in the large vessels, microvasculature, and lymphatics, and how changes in environmental mechanical forces result in changes in metabolism, which ultimately influence cell physiology, cellular memory, and ultimately disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
11
|
Jones DT, Macdonald JH, Sandoo A, Oliver SJ, Rossetti GMK. The deleterious effects of acute hypoxia on microvascular and large vessel endothelial function. Exp Physiol 2021; 106:1699-1709. [PMID: 34036677 DOI: 10.1113/ep089393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim was primarily to determine the effect of hypoxia on microvascular function and secondarily whether superior cardiorespiratory fitness is protective against hypoxia-induced impairment in vascular function. What is the main finding and its importance? Hypoxia reduced endothelium-dependent but not endothelium-independent microvascular function. The extent of impairment was twofold higher in the microcirculation compared with the large blood vessels. This study suggests that individuals with superior cardiorespiratory fitness might preserve microvascular function in hypoxia. These findings highlight the sensitivity of the microvascular circulation to hypoxia. ABSTRACT Hypoxia is associated with diminished bioavailability of the endothelium-derived vasodilator, nitric oxide (NO). Diminished NO bioavailability can have deleterious effects on endothelial function. The endothelium is a heterogeneous tissue; therefore, a comprehensive assessment of endothelial function is crucial to understand the significance of hypoxia-induced endothelial dysfunction. We hypothesized that acute hypoxia would have a deleterious effect on microvascular and large vessel endothelial function. Twenty-nine healthy adults [24 (SD = 4 ) years of age] completed normoxic and hypoxic [inspired O2 fraction = 0.209] trials in this double-blinded, counterbalanced crossover study. After 30 min, we assessed the laser Doppler imaging-determined perfusion response to iontophoresis of ACh as a measure of endothelium-dependent microvascular function and iontophoresis of sodium nitroprusside as a measure of endothelium-independent microvascular function. After 60 min, we assessed brachial flow-mediated dilatation as a measure of large vessel endothelial function. Thirty minutes of hypoxia reduced endothelium-dependent microvascular function determined by the perfusion response to ACh (median difference (x̃∆) = -109% {interquartile range: 542.7}, P < 0.05), but not endothelium-independent microvascular function determined by the perfusion response to sodium nitroprusside (x̃∆ = 69% {interquartile range: 453.7}, P = 0.6). In addition, 60 min of hypoxia reduced allometrically scaled flow-mediated dilatation compared with normoxia ( x ¯ Δ = - 1.19 [95% CI = -1.80, -0.58 (Confidence Intervals)]%, P < 0.001). The decrease in microvascular endothelial function was associated with cardiorespiratory fitness (r = 0.45, P = 0.02). In conclusion, acute exposure to normobaric hypoxia significantly reduced endothelium-dependent vasodilatory capacity in small and large vessels. Collectively, these findings highlight the sensitivity of the microvascular circulation to hypoxic insult, particularly in those with poor cardiorespiratory fitness.
Collapse
Affiliation(s)
- Danial T Jones
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Jamie H Macdonald
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Aamer Sandoo
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Samuel J Oliver
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK
| | - Gabriella M K Rossetti
- School of Sport, Health and Exercise Sciences, College of Human Sciences, Bangor University, Bangor, UK.,Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| |
Collapse
|
12
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
13
|
Sam QH, Ling H, Yew WS, Tan Z, Ravikumar S, Chang MW, Chai LYA. The Divergent Immunomodulatory Effects of Short Chain Fatty Acids and Medium Chain Fatty Acids. Int J Mol Sci 2021; 22:ijms22126453. [PMID: 34208638 PMCID: PMC8234078 DOI: 10.3390/ijms22126453] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 01/22/2023] Open
Abstract
Fatty acids are derived from diet and fermentative processes by the intestinal flora. Two to five carbon chain fatty acids, termed short chain fatty acids (SCFA) are increasingly recognized to play a role in intestinal homeostasis. However, the characteristics of slightly longer 6 to 10 carbon, medium chain fatty acids (MCFA), derived primarily from diet, are less understood. Here, we demonstrated that SCFA and MCFA have divergent immunomodulatory propensities. SCFA down-attenuated host pro-inflammatory IL-1β, IL-6, and TNFα response predominantly through the TLR4 pathway, whereas MCFA augmented inflammation through TLR2. Butyric (C4) and decanoic (C10) acid displayed most potent modulatory effects within the SCFA and MCFA, respectively. Reduction in TRAF3, IRF3 and TRAF6 expression were observed with butyric acid. Decanoic acid induced up-regulation of GPR84 and PPARγ and altered HIF-1α/HIF-2α ratio. These variant immune characteristics of the fatty acids which differ by just several carbon atoms may be attributable to their origins, with SCFA being primarily endogenous and playing a physiological role, and MCFA exogenously from the diet.
Collapse
Affiliation(s)
- Qi Hui Sam
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore; (Q.H.S.); (Z.T.); (S.R.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (H.L.); (W.S.Y.); (M.W.C.)
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Hua Ling
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (H.L.); (W.S.Y.); (M.W.C.)
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Wen Shan Yew
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (H.L.); (W.S.Y.); (M.W.C.)
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Zhaohong Tan
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore; (Q.H.S.); (Z.T.); (S.R.)
| | - Sharada Ravikumar
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore; (Q.H.S.); (Z.T.); (S.R.)
| | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (H.L.); (W.S.Y.); (M.W.C.)
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Louis Yi Ann Chai
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singapore; (Q.H.S.); (Z.T.); (S.R.)
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6779-5555; Fax: +65-6872-4130
| |
Collapse
|
14
|
Shosha E, Fouda AY, Lemtalsi T, Haigh S, Fulton D, Ibrahim A, Al-Shabrawey M, Caldwell RW, Caldwell RB. Endothelial arginase 2 mediates retinal ischemia/reperfusion injury by inducing mitochondrial dysfunction. Mol Metab 2021; 53:101273. [PMID: 34139341 PMCID: PMC8274341 DOI: 10.1016/j.molmet.2021.101273] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Retinal ischemic disease is a major cause of vision loss. Current treatment options are limited to late-stage diseases, and the molecular mechanisms of the initial insult are not fully understood. We have previously shown that the deletion of the mitochondrial arginase isoform, arginase 2 (A2), limits neurovascular injury in models of ischemic retinopathy. Here, we investigated the involvement of A2-mediated alterations in mitochondrial dynamics and function in the pathology. Methods We used wild-type (WT), global A2 knockout (A2KO-) mice, cell-specific A2 knockout mice subjected to retinal ischemia/reperfusion (I/R), and bovine retinal endothelial cells (BRECs) subjected to an oxygen-glucose deprivation/reperfusion (OGD/R) insult. We used western blotting to measure levels of cell stress and death markers and the mitochondrial fragmentation protein, dynamin related protein 1 (Drp1). We also used live cell mitochondrial labeling and Seahorse XF analysis to evaluate mitochondrial fragmentation and function, respectively. Results We found that the global deletion of A2 limited the I/R-induced disruption of retinal layers, fundus abnormalities, and albumin extravasation. The specific deletion of A2 in endothelial cells was protective against I/R-induced neurodegeneration. The OGD/R insult in BRECs increased A2 expression and induced cell stress and cell death, along with decreased mitochondrial respiration, increased Drp1 expression, and mitochondrial fragmentation. The overexpression of A2 in BREC also decreased mitochondrial respiration, promoted increases in the expression of Drp1, mitochondrial fragmentation, and cell stress and resulted in decreased cell survival. In contrast, the overexpression of the cytosolic isoform, arginase 1 (A1), did not affect these parameters. Conclusions This study is the first to show that A2 in endothelial cells mediates retinal ischemic injury through a mechanism involving alterations in mitochondrial dynamics and function. Ischemic retinopathy is a common feature of blinding eye disease. Arginase 2 overexpression in endothelial cells induces mitochondrial dysfunction. Endothelial-specific arginase 2 deletion improves neuronal survival after ischemia. Endothelial cell arginase 2 plays a crucial role in ischemic retinal injury.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Abdelrahman Y Fouda
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Stephen Haigh
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Ahmed Ibrahim
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Wayne State University, Department of Ophthalmology, Visual, and Anatomical Sciences, Department of Pharmacology, Detroit, MI, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed Al-Shabrawey
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Oral Biology, Dental College of Georgia, Augusta, GA, USA
| | - R William Caldwell
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
15
|
Douglass MS, Zhang Y, Kaplowitz MR, Fike CD. L-citrulline increases arginase II protein levels and arginase activity in hypoxic piglet pulmonary artery endothelial cells. Pulm Circ 2021; 11:20458940211006289. [PMID: 33948161 PMCID: PMC8053766 DOI: 10.1177/20458940211006289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/08/2021] [Indexed: 11/15/2022] Open
Abstract
The L-arginine precursor, L-citrulline, re-couples endothelial nitric oxide synthase, increases nitric oxide production, and ameliorates chronic hypoxia-induced pulmonary hypertension in newborn pigs. L-arginine can induce arginase, which, in turn, may diminish nitric oxide production. Our major purpose was to determine if L-citrulline increases arginase activity in hypoxic piglet pulmonary arterial endothelial cells, and if so, concomitantly impacts the ability to increase endothelial nitric oxide synthase re-coupling and nitric oxide production. Piglet pulmonary arterial endothelial cells were cultured in hypoxic conditions with L-citrulline (0-3 mM) and/or the arginase inhibitor S-(2-boronoethyl)-L-cysteine. We measured arginase activity and nitric oxide production. We assessed endothelial nitric oxide synthase coupling by measuring endothelial nitric oxide synthase dimers and monomers. L-citrulline concentrations ≥0.5 mM increased arginase activity in hypoxic pulmonary arterial endothelial cells. L-citrulline concentrations ≥0.1 mM increased nitric oxide production and concentrations ≥0.5 mM elevated endothelial nitric oxide synthase dimer-to-monomer ratios. Co-treatment with L-citrulline and S-(2-boronoethyl)-L-cysteine elevated endothelial nitric oxide synthase dimer-to-monomer ratios more than sole treatment. Despite inducing arginase, L-citrulline increased nitric oxide production and endothelial nitric oxide synthase coupling in hypoxic piglet pulmonary arterial endothelial cells. However, these dose-dependent findings raise the possibility that there could be L-citrulline concentrations that elevate arginase to levels that negate improvements in endothelial nitric oxide synthase dysfunction. Moreover, our findings suggest that combining an arginase inhibitor with L-citrulline merits evaluation as a treatment for chronic hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
| | - Yongmei Zhang
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Mark R Kaplowitz
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Candice D Fike
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Pullamsetti SS, Mamazhakypov A, Weissmann N, Seeger W, Savai R. Hypoxia-inducible factor signaling in pulmonary hypertension. J Clin Invest 2021; 130:5638-5651. [PMID: 32881714 DOI: 10.1172/jci137558] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary artery remodeling that can subsequently culminate in right heart failure and premature death. Emerging evidence suggests that hypoxia-inducible factor (HIF) signaling plays a fundamental and pivotal role in the pathogenesis of PH. This Review summarizes the regulation of HIF isoforms and their impact in various PH subtypes, as well as the elaborate conditional and cell-specific knockout mouse studies that brought the role of this pathway to light. We also discuss the current preclinical status of pan- and isoform-selective HIF inhibitors, and propose new research areas that may facilitate HIF isoform-specific inhibition as a novel therapeutic strategy for PH and right heart failure.
Collapse
Affiliation(s)
- Soni Savai Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, member of the DZL and CPI, Justus Liebig University, Giessen, Germany
| | - Argen Mamazhakypov
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Norbert Weissmann
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, member of the DZL and CPI, Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, member of the DZL and CPI, Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, member of the DZL and CPI, Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
17
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
18
|
Moretto J, Pudlo M, Demougeot C. Human-based evidence for the therapeutic potential of arginase inhibitors in cardiovascular diseases. Drug Discov Today 2020; 26:138-147. [PMID: 33197620 DOI: 10.1016/j.drudis.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Affiliation(s)
- Johnny Moretto
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France.
| | - Marc Pudlo
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
19
|
Mohandas A, Rangasamy J. Nanocurcumin and arginine entrapped injectable chitosan hydrogel for restoration of hypoxia induced endothelial dysfunction. Int J Biol Macromol 2020; 166:471-482. [PMID: 33129903 DOI: 10.1016/j.ijbiomac.2020.10.205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022]
Abstract
Hypoxia is a condition that gradually leads to ischemic damages in organs which is marked by poor tissue perfusion. Depending on the severity of the condition, revascularisation therapies are needed for reducing the risk of organ dysfunction. This study was aimed at developing an injectable nanocurcumin and arginine incorporated chitosan hydrogel (nC/R) that can prevent hypoxia induced endothelial damage. The prepared hydrogel has shear thinning, stable and injectable nature. The (nC and nC/R) hydrogels showed significant antioxidant activity and biodegradation in vitro. The release of curucmin and arginine from the nC/R was found to be higher at acidic pH, which predominates in an ischemic site. To mimic low oxygen environment, an in vitro hypoxic endothelial dysfunction model was developed which showed decreased expressions of phosphorylated eNOS (serine 1177) when compared to the cells cultured in normoxic condition. In vitro tube formation assay demonstrated the protective effect of nC/R towards hypoxia induced reduction of tube width. The nC/R hydrogel was found to enhance phosphorylation of eNOS at serine 1177 site in cultured endothelial cells subjected to hypoxia. Therefore, nC/R hydrogel could effectively deliver both curcumin and arginine and therapeutically reduce the effect of hypoxia induced endothelial dysfunction.
Collapse
Affiliation(s)
- Annapoorna Mohandas
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682 041, India
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682 041, India.
| |
Collapse
|
20
|
Sanhueza C, Bennett JC, Valenzuela-Valderrama M, Contreras P, Lobos-González L, Campos A, Wehinger S, Lladser Á, Kiessling R, Leyton L, Quest AF. Caveolin-1-Mediated Tumor Suppression Is Linked to Reduced HIF1α S-Nitrosylation and Transcriptional Activity in Hypoxia. Cancers (Basel) 2020; 12:cancers12092349. [PMID: 32825247 PMCID: PMC7565942 DOI: 10.3390/cancers12092349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Caveolin-1 (CAV1) is a well-established nitric oxide synthase inhibitor, whose function as a tumor suppressor is favored by, but not entirely dependent on, the presence of E-cadherin. Tumors are frequently hypoxic and the activation of the hypoxia-inducible factor-1α (HIF1α) promotes tumor growth. HIF1α is regulated by several post-translational modifications, including S-nitrosylation. Here, we evaluate the mechanisms underlying tumor suppression by CAV1 in cancer cells lacking E-cadherin in hypoxia. Our main findings are that CAV1 reduced HIF activity and Vascular Endothelial Growth Factor expression in vitro and in vivo. This effect was neither due to reduced HIF1α protein stability or reduced nuclear translocation. Instead, HIF1α S-nitrosylation observed in hypoxia was diminished by the presence of CAV1, and nitric oxide synthase (NOS) inhibition by Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) reduced HIF1α transcriptional activity in cells to the same extent as observed upon CAV1 expression. Additionally, arginase inhibition by (S)-(2-Boronoethyl)-L-cysteine (BEC) partially rescued cells from the CAV1-mediated suppression of HIF1α transcriptional activity. In vivo, CAV1-mediated tumor suppression was dependent on NOS activity. In summary, CAV1-dependent tumor suppression in the absence of E-cadherin is linked to reduced HIF1α transcriptional activity via diminished NOS-mediated HIF1α S-nitrosylation.
Collapse
Affiliation(s)
- Carlos Sanhueza
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Instituto Oncológico Fundación Arturo López Pérez, Santiago 7500921, Chile
| | - Jimena Castillo Bennett
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
| | - Manuel Valenzuela-Valderrama
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
- Laboratorio de Microbiología Celular, Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8320000, Chile
| | - Pamela Contreras
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana Universidad Del Desarrollo, Santiago 7710162, Chile
| | - América Campos
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
| | - Sergio Wehinger
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile
| | - Álvaro Lladser
- Laboratory of Immunoncology, Fundación Ciencia & Vida; Facultad de Medicina y Ciencia, Universidad San Sebastián; Santiago 7780272, Chile;
| | - Rolf Kiessling
- Immune and Gene Therapy Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 17164 Stockholm, Sweden;
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
| | - Andrew F.G. Quest
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.S.); (J.C.B.); (P.C.); (A.C.); (S.W.); (L.L.)
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380000, Chile; (M.V.-V.); (L.L.-G.)
- Correspondence: ; Tel.: +56-2-29786832
| |
Collapse
|
21
|
Wernly B, Pernow J, Kelm M, Jung C. The role of arginase in the microcirculation in cardiovascular disease. Clin Hemorheol Microcirc 2020; 74:79-92. [PMID: 31743994 DOI: 10.3233/ch-199237] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the microcirculation, the exchange of nutrients, water, gas, hormones, and waste takes place, and it is divided into the three main sections arterioles, capillaries, and venules. Disturbances in the microcirculation can be measured using surrogate parameters or be visualized either indirectly or directly.Arginase is a manganese metalloenzyme hydrolyzing L-arginine to urea and L-ornithine. It is located in different cell types, including vascular cells, but also in circulating cells such as red blood cells. A variety of pro-inflammatory factors, as well as interleukins, stimulate increased arginase expression. An increase in arginase activity consequently leads to a consumption of L-arginine needed for nitric oxide (NO) production by endothelial NO synthase. A vast body of evidence convincingly showed that increased arginase activity is associated with endothelial dysfunction in larger vessels of the vascular tree. Of note, arginase also influences the microcirculation. Arginase inhibition leads to an increase in the bioavailability of NO and reduces superoxide levels, resulting in improved endothelial function. Arginase inhibition might, therefore, be a potent treatment strategy in cardiovascular medicine. Recently, red blood cells emerged as an influential player in the development from increased arginase activity to endothelial dysfunction. As red blood cells directly interact with the microcirculation in gas exchange, this could constitute a potential link between arginase activity, endothelial dysfunction and microcirculatory disturbances.The aim of this review is to summarize recent findings revealing the role of arginase in regulating vascular function with particular emphasis on the microcirculation.
Collapse
Affiliation(s)
- Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Düsseldorf, Düsseldorf, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Mahdi A, Kövamees O, Pernow J. Improvement in endothelial function in cardiovascular disease - Is arginase the target? Int J Cardiol 2019; 301:207-214. [PMID: 31785959 DOI: 10.1016/j.ijcard.2019.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/26/2019] [Accepted: 11/04/2019] [Indexed: 01/30/2023]
Abstract
Endothelial dysfunction represents an early change in the vascular wall in areas prone to atherosclerotic plaque formation and is present in association with several risk factors for cardiovascular disease. The underlying mechanisms behind endothelial dysfunction are multifactorial and complex. Arginase has emerged as a key player in the regulation of endothelial integrity by the ability of reciprocally inhibits nitric oxide formation and promoting oxidative stress. A chain of evidence suggest that arginase is implicated in the pathogenesis underlying endothelial dysfunction induced by several cardiovascular risk factors and established cardiovascular disease including diabetes, hypercholesteremia, ischemia/reperfusion, atherosclerosis, obesity, ageing and hypertension. Recent data has unveiled a key role of arginase as one of the key mechanisms underlying endothelial dysfunction in diabetes and may serve as a potential therapeutic target in previously overlooked compartments including red blood cells. The current review is devoted to discuss arginase as a key mediator in endothelial dysfunction and the potential for therapeutic possibilities to target this enzyme in various diseases, especially type 2 diabetes, atherosclerosis and ischemia/reperfusion with focus on translational and clinical aspects. Moreover, approaches of how and in which patient group(s) arginase may be targeted in future clinical trials are discussed.
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
23
|
Zemskov EA, Lu Q, Ornatowski W, Klinger CN, Desai AA, Maltepe E, Yuan JXJ, Wang T, Fineman JR, Black SM. Biomechanical Forces and Oxidative Stress: Implications for Pulmonary Vascular Disease. Antioxid Redox Signal 2019; 31:819-842. [PMID: 30623676 PMCID: PMC6751394 DOI: 10.1089/ars.2018.7720] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christina N Klinger
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, Arizona
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
24
|
Liang X, Arullampalam P, Yang Z, Ming XF. Hypoxia Enhances Endothelial Intercellular Adhesion Molecule 1 Protein Level Through Upregulation of Arginase Type II and Mitochondrial Oxidative Stress. Front Physiol 2019; 10:1003. [PMID: 31474872 PMCID: PMC6702258 DOI: 10.3389/fphys.2019.01003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/19/2019] [Indexed: 02/04/2023] Open
Abstract
Hypoxia plays a crucial role in the pathogenesis of cardiovascular diseases. Mitochondrial enzyme arginase type II (Arg-II) is reported to lead to endothelial dysfunction and enhance the expression of endothelial inflammatory adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). In this study, we investigate the role of Arg-II in hypoxia-induced endothelial activation and the potential underlying mechanisms. Exposure of the human endothelial cells to hypoxia induced a time-dependent increase in Arg-II, HIF1α, HIF2α, and ICAM-1 protein level, whereas no change in the protein level of VCAM-1 and E-selectin was observed. Similar effects were obtained in cells treated with a hypoxia mimetic Dimethyloxaloylglycine (DMOG). Silencing HIF1α, but not HIF2α, reversed hypoxia-induced upregulation of Arg-II. Moreover, silencing Arg-II prevented the ICAM-1 upregulation induced by hypoxia or DMOG. Furthermore, the endothelial cells incubated under hypoxic condition or treated with DMOG or hypoxia enhanced monocyte adhesion, which was inhibited by silencing Arg-II. Lastly, silencing Arg-II prevented hypoxia-induced mitochondrial superoxide production in endothelial cells, and hypoxia-induced ICAM-1 upregulation was reversed by mitochondrial electron transport inhibitor rotenone. These data demonstrate that hypoxia enhances ICAM-1 protein level and monocyte-endothelial interaction through HIF1α-mediated increase in Arg-II protein level on leading to increased mitochondrial reactive oxygen species production. These effects of hypoxia on endothelial cells may play a key role in cardiovascular diseases. Our results suggest that Arg-II could be a promising therapeutic target to prevent hypoxia-induced vascular damage/dysfunction.
Collapse
Affiliation(s)
- Xiujie Liang
- Laboratory of Cardiovascular and Aging Research, Medicine Section, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Prakash Arullampalam
- Laboratory of Cardiovascular and Aging Research, Medicine Section, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Zhihong Yang
- Laboratory of Cardiovascular and Aging Research, Medicine Section, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Xiu-Fen Ming
- Laboratory of Cardiovascular and Aging Research, Medicine Section, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
25
|
Serman Y, Fuentealba RA, Pasten C, Rocco J, Ko BCB, Carrión F, Irarrázabal CE. Emerging new role of NFAT5 in inducible nitric oxide synthase in response to hypoxia in mouse embryonic fibroblast cells. Am J Physiol Cell Physiol 2019; 317:C31-C38. [PMID: 31067085 DOI: 10.1152/ajpcell.00054.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously described the protective role of the nuclear factor of activated T cells 5 (NFAT5) during hypoxia. Alternatively, inducible nitric oxide synthase (iNOS) is also induced by hypoxia. Some evidence indicates that NFAT5 is essential for the expression of iNOS in Toll-like receptor-stimulated macrophages and that iNOS inhibition increases NFAT5 expression in renal ischemia-reperfusion. Here we studied potential NFAT5 target genes stimulated by hypoxia in mouse embryonic fibroblast (MEF) cells. We used three types of MEF cells associated with NFAT5 gene: NFAT5 wild type (MEF-NFAT5+/+), NFAT5 knockout (MEF-NFAT5-/-), and NFAT5 dominant-negative (MEF-NFAT5Δ/Δ) cells. MEF cells were exposed to 21% or 1% O2 in a time course curve of 48 h. We found that, in MEF-NFAT5+/+ cells exposed to 1% O2, NFAT5 was upregulated and translocated into the nuclei, and its transactivation domain activity was induced, concomitant with iNOS, aquaporin 1 (AQP-1), and urea transporter 1 (UTA-1) upregulation. Interestingly, in MEF-NFAT5-/- or MEF-NFAT5Δ/Δ cells, the basal levels of iNOS and AQP-1 expression were strongly downregulated, but not for UTA-1. The upregulation of AQP-1, UTA-1, and iNOS by hypoxia was blocked in both NFAT5-mutated cells. The iNOS induction by hypoxia was recovered in MEF-NFAT5-/- MEF cells, when recombinant NFAT5 protein expression was reconstituted, but not in MEF-NFAT5Δ/Δ cells, confirming the dominant-negative effect of MEF-NFAT5Δ/Δ cells. We did not see the rescue effect on AQP-1 expression. This work provides novel and relevant information about the signaling pathway of NFAT5 during responses to oxygen depletion in mammalian cells and suggests that the expression of iNOS induced by hypoxia is dependent on NFAT5.
Collapse
Affiliation(s)
- Yair Serman
- Laboratorio de Fisiología Integrativa y Molecular, Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes , Santiago , Chile
| | - Rodrigo A Fuentealba
- Laboratorio de Fisiología Integrativa y Molecular, Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes , Santiago , Chile
| | - Consuelo Pasten
- Laboratorio de Fisiología Integrativa y Molecular, Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes , Santiago , Chile
| | - Jocelyn Rocco
- Laboratorio de Fisiología Integrativa y Molecular, Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes , Santiago , Chile
| | - Ben C B Ko
- Department of Applied Biology and Chemical Technology, Polytechnic University of Hong Kong, Hong Kong, China
| | - Flavio Carrión
- Programa de Inmunología Traslacional, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo , Santiago , Chile
| | - Carlos E Irarrázabal
- Laboratorio de Fisiología Integrativa y Molecular, Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes , Santiago , Chile
| |
Collapse
|
26
|
Ng KP, Manjeri A, Lee LM, Chan ZE, Tan CY, Tan QD, Majeed A, Lee KL, Chuah C, Suda T, Ong ST. The arginase inhibitor Nω-hydroxy-nor-arginine (nor-NOHA) induces apoptosis in leukemic cells specifically under hypoxic conditions but CRISPR/Cas9 excludes arginase 2 (ARG2) as the functional target. PLoS One 2018; 13:e0205254. [PMID: 30307989 PMCID: PMC6181325 DOI: 10.1371/journal.pone.0205254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/23/2018] [Indexed: 01/10/2023] Open
Abstract
Cancer cells, including in chronic myeloid leukemia (CML), depend on the hypoxic response to persist in hosts and evade therapy. Accordingly, there is significant interest in drugging cancer-specific hypoxic responses. However, a major challenge in leukemia is identifying differential and druggable hypoxic responses between leukemic and normal cells. Previously, we found that arginase 2 (ARG2), an enzyme of the urea cycle, is overexpressed in CML but not normal progenitors. ARG2 is a target of the hypoxia inducible factors (HIF1−α and HIF2−α), and is required for the generation of polyamines which are required for cell growth. We therefore explored if the clinically-tested arginase inhibitor Nω−hydroxy−nor−arginine (nor−NOHA) would be effective against leukemic cells under hypoxic conditions. Remarkably, nor−NOHA effectively induced apoptosis in ARG2-expressing cells under hypoxia but not normoxia. Co-treatment with nor−NOHA overcame hypoxia-mediated resistance towards BCR−ABL1 kinase inhibitors. While nor−NOHA itself is promising in targeting the leukemia hypoxic response, we unexpectedly found that its anti-leukemic activity was independent of ARG2 inhibition. Genetic ablation of ARG2 using CRISPR/Cas9 had no effect on the viability of leukemic cells and their sensitivity towards nor−NOHA. This discrepancy was further evidenced by the distinct effects of ARG2 knockouts and nor−NOHA on cellular respiration. In conclusion, we show that nor−NOHA has significant but off-target anti-leukemic activity among ARG2-expressing hypoxic cells. Since nor−NOHA has been employed in clinical trials, and is widely used in studies on endothelial dysfunction, immunosuppression and metabolism, the diverse biological effects of nor−NOHA must be cautiously evaluated before attributing its activity to ARG inhibition.
Collapse
Affiliation(s)
- King Pan Ng
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Aditi Manjeri
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Lin Ming Lee
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Zhu En Chan
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chin Yee Tan
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Qiancheng Darren Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - A'Qilah Majeed
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kian Leong Lee
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Charles Chuah
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,International Research Center for Medical Sciences, Kumamoto University, Japan
| | - S Tiong Ong
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Department of Haematology, Singapore General Hospital, Singapore, Singapore.,Department of Medical Oncology, National Cancer Centre, Singapore, Singapore.,Department of Medicine, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
27
|
Krause BJ, Casanello P, Dias AC, Arias P, Velarde V, Arenas GA, Preite MD, Iturriaga R. Chronic Intermittent Hypoxia-Induced Vascular Dysfunction in Rats is Reverted by N-Acetylcysteine Supplementation and Arginase Inhibition. Front Physiol 2018; 9:901. [PMID: 30087615 PMCID: PMC6066978 DOI: 10.3389/fphys.2018.00901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 01/07/2023] Open
Abstract
Chronic intermittent hypoxia (CIH), the main attribute of obstructive sleep apnea (OSA), produces oxidative stress, endothelial dysfunction, and hypertension. Nitric oxide (NO) plays a critical role in controlling the vasomotor tone. The NO level depends on the L-arginine level, which can be reduced by arginase enzymatic activity, and its reaction with the superoxide radical to produce peroxynitrite. Accordingly, we hypothesized whether a combination of an arginase inhibitor and an antioxidant may restore the endothelial function and reduced arterial blood pressure (BP) in CIH-induced hypertensive rats. Male Sprague-Dawley rats 200 g were exposed either to CIH (5% O2, 12 times/h 8 h/day) or sham condition for 35 days. BP was continuously measured by radio-telemetry in conscious animals. After 14 days, rats were treated with 2(S)-amino-6-boronohexanoic acid (ABH 400 μg/kg day, osmotic pump), N-acetylcysteine (NAC 100 mg/kg day, drinking water), or the combination of both drugs until day 35. At the end of the experiments, external carotid and femoral arteries were isolated to determine vasoactive contractile responses induced by KCL and acetylcholine (ACh) with wire-myography. CIH-induced hypertension (~8 mmHg) was reverted by ABH, NAC, and ABH/NAC administration. Carotid arteries from CIH-treated rats showed higher contraction induced by KCl (3.4 ± 0.4 vs. 2.4 ± 0.2 N/m2) and diminished vasorelaxation elicits by ACh compared to sham rats (12.8 ± 1.5 vs. 30.5 ± 4.6%). ABH reverted the increased contraction (2.5 ± 0.2 N/m2) and the reduced vasorelaxation induced by ACh in carotid arteries from CIH-rats (38.1 ± 4.9%). However, NAC failed to revert the enhanced vasocontraction (3.9 ± 0.6 N/m2) induced by KCl and the diminished ACh-induced vasorelaxation in carotid arteries (10.7 ± 0.8%). Femoral arteries from CIH rats showed an increased contractile response, an effect partially reverted by ABH, but completely reverted by NAC and ABH/NAC. The impaired endothelial-dependent relaxation in femoral arteries from CIH rats was reverted by ABH and ABH/NAC. In addition, ABH/NAC at high doses had no effect on liver and kidney gross morphology and biochemical parameters. Thus, although ABH, and NAC alone and the combination of ABH/NAC were able to normalize the elevated BP, only the combined treatment of ABH/NAC normalized the vascular reactivity and the systemic oxidative stress in CIH-treated rats.
Collapse
Affiliation(s)
- Bernardo J Krause
- Division of Pediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Casanello
- Division of Pediatrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Division of Obstetrics & Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ana C Dias
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Arias
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victoria Velarde
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - German A Arenas
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo D Preite
- Departamento de Química Orgánica, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
28
|
Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: A Multifaceted Enzyme Important in Health and Disease. Physiol Rev 2018; 98:641-665. [PMID: 29412048 PMCID: PMC5966718 DOI: 10.1152/physrev.00037.2016] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
The arginase enzyme developed in early life forms and was maintained during evolution. As the last step in the urea cycle, arginase cleaves l-arginine to form urea and l-ornithine. The urea cycle provides protection against excess ammonia, while l-ornithine is needed for cell proliferation, collagen formation, and other physiological functions. In mammals, increases in arginase activity have been linked to dysfunction and pathologies of the cardiovascular system, kidney, and central nervous system and also to dysfunction of the immune system and cancer. Two important aspects of the excessive activity of arginase may be involved in diseases. First, overly active arginase can reduce the supply of l-arginine needed for the production of nitric oxide (NO) by NO synthase. Second, too much l-ornithine can lead to structural problems in the vasculature, neuronal toxicity, and abnormal growth of tumor cells. Seminal studies have demonstrated that increased formation of reactive oxygen species and key inflammatory mediators promote this pathological elevation of arginase activity. Here, we review the involvement of arginase in diseases affecting the cardiovascular, renal, and central nervous system and cancer and discuss the value of therapies targeting the elevated activity of arginase.
Collapse
Affiliation(s)
- R William Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Paulo C Rodriguez
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - S Priya Narayanan
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Ruth B Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| |
Collapse
|
29
|
Pandey D, Nomura Y, Rossberg MC, Hori D, Bhatta A, Keceli G, Leucker T, Santhanam L, Shimoda LA, Berkowitz D, Romer L. Hypoxia Triggers SENP1 (Sentrin-Specific Protease 1) Modulation of KLF15 (Kruppel-Like Factor 15) and Transcriptional Regulation of Arg2 (Arginase 2) in Pulmonary Endothelium. Arterioscler Thromb Vasc Biol 2018; 38:913-926. [PMID: 29472234 DOI: 10.1161/atvbaha.117.310660] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/06/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE KLF15 (Kruppel-like factor 15) has recently been shown to suppress activation of proinflammatory processes that contribute to atherogenesis in vascular smooth muscle, however, the role of KLF15 in vascular endothelial function is unknown. Arginase mediates inflammatory vasculopathy and vascular injury in pulmonary hypertension. Here, we tested the hypothesis that KLF15 is a critical regulator of hypoxia-induced Arg2 (arginase 2) transcription in human pulmonary microvascular endothelial cells (HPMEC). APPROACH AND RESULTS Quiescent HPMEC express ample amounts of full-length KLF15. HPMECs exposed to 24 hours of hypoxia exhibited a marked decrease in KLF15 protein levels and a reciprocal increase in Arg2 protein and mRNA. Chromatin immunoprecipitation indicated direct binding of KLF15 to the Arg2 promoter, which was relieved with HPMEC exposure to hypoxia. Furthermore, overexpression of KLF15 in HPMEC reversed hypoxia-induced augmentation of Arg2 abundance and arginase activity and rescued nitric oxide (NO) production. Ectopic KLF15 also reversed hypoxia-induced endothelium-mediated vasodilatation in isolated rat pulmonary artery rings. Mechanisms by which hypoxia regulates KLF15 abundance, stability, and compartmentalization to the nucleus in HPMEC were then investigated. Hypoxia triggered deSUMOylation of KLF15 by SENP1 (sentrin-specific protease 1), and translocation of KLF15 from nucleus to cytoplasm. CONCLUSIONS KLF15 is a critical regulator of pulmonary endothelial homeostasis via repression of endothelial Arg2 expression. KLF15 abundance and nuclear compartmentalization are regulated by SUMOylation/deSUMOylation-a hypoxia-sensitive process that is controlled by SENP1. Strategies including overexpression of KLF15 or inhibition of SENP1 may represent novel therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Deepesh Pandey
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD.
| | - Yohei Nomura
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Max C Rossberg
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Daijiro Hori
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anil Bhatta
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Gizem Keceli
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Thorsten Leucker
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lakshmi Santhanam
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Larissa A Shimoda
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Dan Berkowitz
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lewis Romer
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
30
|
Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial Cell Metabolism. Physiol Rev 2018; 98:3-58. [PMID: 29167330 PMCID: PMC5866357 DOI: 10.1152/physrev.00001.2017] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
31
|
Brown MB, Kempf A, Collins CM, Long GM, Owens M, Gupta S, Hellman Y, Wong V, Farber M, Lahm T. A prescribed walking regimen plus arginine supplementation improves function and quality of life for patients with pulmonary arterial hypertension: a pilot study. Pulm Circ 2017; 8:2045893217743966. [PMID: 29199900 PMCID: PMC5731727 DOI: 10.1177/2045893217743966] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current evidence suggests that exercise training is beneficial in pulmonary arterial hypertension (PAH). Unfortunately, the standard supervised, hospital-based programs limit patient accessibility to this important intervention. Our proof-of-concept study aimed to provide insight into the usefulness of a prescribed walking regimen along with arginine supplementation to improve outcomes for patients with PAH. Twelve PAH patients (all women) in New York Heart Association (NYHA) functional class (FC) II (n = 7) or III (n = 5) and in stable condition for ≥ 3 months were enrolled. Patients performed home- and fitness-center- based walking at 65-75% heart rate (HR) reserve for 45 min, six sessions/week for 12 weeks. Concomitant L-arginine supplementation (6000 mg/day) was provided to maximize beneficial endothelial training adaptations. Cardiopulmonary exercise testing, 6-min walk testing (6MWT), echocardiography, laboratory studies, and quality of life (QoL) survey (SF-36) were performed at baseline and 12 weeks. Eleven patients completed the study (72 session adherence rate = 96 ± 3%). Objective improvement was demonstrated by the 6MWT distance (increased by 40 ± 13 m, P = 0.01), VO2max (increased by 2 ± 0.7 mL/kg/min, P = 0.02), time-to-VO2max (increased by 2.5 ± 0.6 min, P = 0.001), VO2 at anaerobic threshold (increased by 1.3 ± 0.5 mL/kg/min, P = 0.04), HR recovery (reduced by 68 ± 23% in slope, P = 0.01), and SF-36 subscales of Physical Functioning and Energy/Fatigue (increased by 70 ± 34% and 74 ± 34%, respectively, P < 0.05). No adverse events occurred, and right ventricular function and brain natriuretic peptide levels remained stable, suggesting safety of the intervention. This proof-of-concept study indicates that a simple walking regimen with arginine supplementation is a safe and efficacious intervention for clinically stable PAH patients, with gains in objective function and QoL measures. Further investigation in a randomized controlled trial is warranted.
Collapse
Affiliation(s)
- Mary Beth Brown
- 1 Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA.,2 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Attie Kempf
- 1 Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA
| | - Catherine M Collins
- 1 Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA
| | - Gary M Long
- 1 Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA
| | - Matthew Owens
- 1 Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA
| | - Shikha Gupta
- 2 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yaron Hellman
- 3 12250 Division of Cardiology, Department of Medicine, Indiana University School of Medicine , Indianapolis, IN, USA
| | - Vincent Wong
- 3 12250 Division of Cardiology, Department of Medicine, Indiana University School of Medicine , Indianapolis, IN, USA
| | - Mark Farber
- 2 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tim Lahm
- 2 Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,4 Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
32
|
Bhatta A, Yao L, Xu Z, Toque HA, Chen J, Atawia RT, Fouda AY, Bagi Z, Lucas R, Caldwell RB, Caldwell RW. Obesity-induced vascular dysfunction and arterial stiffening requires endothelial cell arginase 1. Cardiovasc Res 2017; 113:1664-1676. [PMID: 29048462 PMCID: PMC6410953 DOI: 10.1093/cvr/cvx164] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/16/2017] [Accepted: 08/09/2017] [Indexed: 02/04/2023] Open
Abstract
AIMS Elevation of arginase activity has been linked to vascular dysfunction in diabetes and hypertension by a mechanism involving decreased nitric oxide (NO) bioavailability due to L-arginine depletion. Excessive arginase activity also can drive L-arginine metabolism towards the production of ornithine, polyamines, and proline, promoting proliferation of vascular smooth muscle cells and collagen formation, leading to perivascular fibrosis. We hypothesized that there is a specific involvement of arginase 1 expression within the vascular endothelial cells in this pathology. METHODS AND RESULTS To test this proposition, we used models of type 2 diabetes and metabolic syndrome. Studies were performed using wild type (WT), endothelial-specific arginase 1 knockout (EC-A1-/-) and littermate controls(A1con) mice fed high fat-high sucrose (HFHS) or normal diet (ND) for 6 months and isolated vessels exposed to palmitate-high glucose (PA/HG) media. Some WT mice or isolated vessels were treated with an arginase inhibitor, ABH [2-(S)-amino-6-boronohexanoic acid. In WT mice, the HFHS diet promoted increases in body weight, fasting blood glucose, and post-prandial insulin levels along with arterial stiffening and fibrosis, elevated blood pressure, decreased plasma levels of L-arginine, and elevated L-ornithine. The HFHS diet or PA/HG treatment also induced increases in vascular arginase activity along with oxidative stress, reduced vascular NO levels, and impaired endothelial-dependent vasorelaxation. All of these effects except obesity and hypercholesterolemia were prevented or significantly reduced by endothelial-specific deletion of arginase 1 or ABH treatment. CONCLUSION Vascular dysfunctions in diet-induced obesity are prevented by deletion of arginase 1 in vascular endothelial cells or arginase inhibition. These findings indicate that upregulation of arginase 1 expression/activity in vascular endothelial cells has an integral role in diet-induced cardiovascular dysfunction and metabolic syndrome.
Collapse
MESH Headings
- Animals
- Arginase/antagonists & inhibitors
- Arginase/genetics
- Arginase/metabolism
- Arginine/blood
- Blood Glucose/metabolism
- Blood Pressure
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/prevention & control
- Diet, High-Fat
- Dietary Sucrose
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/pharmacology
- Fibrosis
- Genetic Predisposition to Disease
- Insulin/blood
- Male
- Metabolic Syndrome/enzymology
- Metabolic Syndrome/genetics
- Metabolic Syndrome/physiopathology
- Metabolic Syndrome/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Obesity/drug therapy
- Obesity/enzymology
- Obesity/genetics
- Obesity/physiopathology
- Ornithine/blood
- Oxidative Stress
- Phenotype
- Signal Transduction
- Vascular Diseases/enzymology
- Vascular Diseases/genetics
- Vascular Diseases/physiopathology
- Vascular Diseases/prevention & control
- Vascular Stiffness/drug effects
- Vasodilation
Collapse
Affiliation(s)
- Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- School of Pharmaceutical Sciences, South China Research Centre for
Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR
China
| | - Zhimin Xu
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Reem T. Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Abdelrahman Y. Fouda
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Zsolt Bagi
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Ruth B. Caldwell
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta
University, Augusta, GA 30912, USA
- Veterans Administration Medical Centre, Augusta, GA 30912, USA
| | - Robert W. Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| |
Collapse
|
33
|
Arginase Inhibition Reverses Monocrotaline-Induced Pulmonary Hypertension. Int J Mol Sci 2017; 18:ijms18081609. [PMID: 28757567 PMCID: PMC5578001 DOI: 10.3390/ijms18081609] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/13/2017] [Accepted: 07/20/2017] [Indexed: 01/19/2023] Open
Abstract
Pulmonary hypertension (PH) is a heterogeneous disorder associated with a poor prognosis. Thus, the development of novel treatment strategies is of great interest. The enzyme arginase (Arg) is emerging as important player in PH development. The aim of the current study was to determine the expression of ArgI and ArgII as well as the effects of Arg inhibition in a rat model of PH. PH was induced in 35 Sprague–Dawley rats by monocrotaline (MCT, 60 mg/kg as single-dose). There were three experimental groups: sham-treated controls (control group, n = 11), MCT-induced PH (MCT group, n = 11) and MCT-induced PH treated with the Arg inhibitor Nω-hydroxy-nor-l-arginine (nor-NOHA; MCT/NorNoha group, n = 13). ArgI and ArgII expression was determined by immunohistochemistry and Western blot. Right ventricular systolic pressure (RVPsys) was measured and lung tissue remodeling was determined. Induction of PH resulted in an increase in RVPsys (81 ± 16 mmHg) compared to the control group (41 ± 15 mmHg, p = 0.002) accompanied by a significant elevation of histological sum-score (8.2 ± 2.4 in the MCT compared to 1.6 ± 1.6 in the control group, p < 0.001). Both, ArgI and ArgII were relevantly expressed in lung tissue and there was a significant increase in the MCT compared to the control group (p < 0.01). Arg inhibition resulted in a significant reduction of RVPsys to 52 ± 19 mmHg (p = 0.006) and histological sum-score to 5.8 ± 1.4 compared to the MCT group (p = 0.022). PH leads to increased expression of Arg. Arg inhibition leads to reduction of RVPsys and diminished lung tissue remodeling and therefore represents a potential treatment strategy in PH.
Collapse
|
34
|
Hussain A, Suleiman MS, George SJ, Loubani M, Morice A. Hypoxic Pulmonary Vasoconstriction in Humans: Tale or Myth. Open Cardiovasc Med J 2017; 11:1-13. [PMID: 28217180 PMCID: PMC5301302 DOI: 10.2174/1874192401711010001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/02/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
Hypoxic Pulmonary vasoconstriction (HPV) describes the physiological adaptive process of lungs to preserves systemic oxygenation. It has clinical implications in the development of pulmonary hypertension which impacts on outcomes of patients undergoing cardiothoracic surgery. This review examines both acute and chronic hypoxic vasoconstriction focusing on the distinct clinical implications and highlights the role of calcium and mitochondria in acute versus the role of reactive oxygen species and Rho GTPases in chronic HPV. Furthermore it identifies gaps of knowledge and need for further research in humans to clearly define this phenomenon and the underlying mechanism.
Collapse
Affiliation(s)
- A Hussain
- Department of Cardiothoracic Surgery, Castle Hill Hospital, Castle Road, Cottingham, HU16 5JQ, UK
| | - M S Suleiman
- School of Clinical Sciences, Bristol Royal Infirmary, Marlborough Street, Bristol, BS2 8HW, UK
| | - S J George
- School of Clinical Sciences, Bristol Royal Infirmary, Marlborough Street, Bristol, BS2 8HW, UK
| | - M Loubani
- Department of Cardiothoracic Surgery, Castle Hill Hospital, Castle Road, Cottingham, HU16 5JQ, UK
| | - A Morice
- Department of Respiratory Medicine, Castle Hill Hospital, Castle Road, Cottingham, HU16 5JQ, UK
| |
Collapse
|
35
|
Wu J, Ke X, Fu W, Gao X, Zhang H, Wang W, Ma N, Zhao M, Hao X, Zhang Z. Inhibition of Hypoxia-Induced Retinal Angiogenesis by Specnuezhenide, an Effective Constituent of Ligustrum lucidum Ait., through Suppression of the HIF-1α/VEGF Signaling Pathway. Molecules 2016; 21:molecules21121756. [PMID: 28009852 PMCID: PMC6272965 DOI: 10.3390/molecules21121756] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/10/2016] [Accepted: 12/17/2016] [Indexed: 11/30/2022] Open
Abstract
Specnuezhenide (SPN), one of the main ingredients of Chinese medicine “Nü-zhen-zi”, has anti-angiogenic and vision improvement effects. However, studies of its effect on retinal neovascularization are limited so far. In the present study, we established a vascular endothelial growth factor A (VEGFA) secretion model of human acute retinal pigment epithelial-19 (ARPE-19) cells by exposure of 150 μM CoCl2 to the cells and determined the VEGFA concentrations, the mRNA expressions of VEGFA, hypoxia inducible factor-1α (HIF-1α) & prolyl hydroxylases 2 (PHD-2), and the protein expressions of HIF-1α and PHD-2 after treatment of 3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole (YC-1, 1.0 μg/mL) or SPN (0.2, 1.0 and 5.0 μg/mL). Furthermore, rat pups with retinopathy were treated with SPN (5.0 and 10.0 mg/kg) in an 80% oxygen atmosphere and the retinal avascular areas were assessed through visualization using infusion of ADPase and H&E stains. The results showed that SPN inhibited VEGFA secretion by ARPE-19 cells under hypoxia condition, down-regulated the mRNA expressions of VEGFA and PHD-2 slightly, and the protein expressions of VEGFA, HIF-1α and PHD-2 significantly in vitro. SPN also prevented hypoxia-induced retinal neovascularization in a rat model of oxygen-induced retinopathy in vivo. These results indicate that SPN ameliorates retinal neovascularization through inhibition of HIF-1α/VEGF signaling pathway. Therefore, SPN has the potential to be developed as an agent for the prevention and treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Jianming Wu
- Laboratory of Chinese Materia Medica, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
- Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xiao Ke
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Wei Fu
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Xiaoping Gao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Hongcheng Zhang
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Wei Wang
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Na Ma
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Manxi Zhao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Xiaofeng Hao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu 610036, Sichuan, China.
| | - Zhirong Zhang
- Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
36
|
Wu J, Ke X, Wang W, Zhang H, Ma N, Fu W, Zhao M, Gao X, Hao X, Zhang Z. Aloe-emodin suppresses hypoxia-induced retinal angiogenesis via inhibition of HIF-1α/VEGF pathway. Int J Biol Sci 2016; 12:1363-1371. [PMID: 27877088 PMCID: PMC5118782 DOI: 10.7150/ijbs.16334] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/22/2016] [Indexed: 11/05/2022] Open
Abstract
Background: Aloe-emodin (AE) has been reported to possess the antiangiogenic effect on laser induced choroidal neovascularization. AE inhibits the vessel formation in the zebrafish embryos. However, it is still unclear whether AE can alleviate neovascularization. Here, we investigated the inhibitory effect of AE on the hypoxia-induced retinal neovascularization and the possible mechanisms. Methods: We established a vascular endothelial growth factor (VEGF) secretion model under chemical induced hypoxia by exposure of 150 µM CoCl2 to the ARPE-19 cells, then treated the cells with different concentrations of AE (0.2, 1.0 and 5.0 µg/mL) or a special hypoxia-inducible factor 1α (HIF-1α) inhibitor [3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole, YC-1, 1.0 µg/mL]. The cellular supernatants were collected 48 h later to measure the VEGFA concentrations by human VEGFA enzyme-linked immunosorbent assay (ELISA) kits, the mRNA expressions of VEGFA, HIF-1α and prolyl hydroxylase-2 (PHD-2) by quantitative reverse transcription-PCR (qRT-PCR) and the protein expressions of HIF-1α and PHD-2 by Western blots. For in vivo study, the rat pups with oxygen-induced retinopathy were treated with Conbercept ophthalmic injection (1.0 mg/kg) or AE (5.0 and 10.0 mg/kg) for five days, then the retinal avascular areas were assessed via visualization of the retinal vasculature with ADPase and hematoxylin & eosin (H&E) stains. Results: AE inhibits the VEGFA secretion of ARPE-19 cells under hypoxia condition, decreases the mRNA expressions of VEGFA and PHD-2 and the protein expressions of VEGFA, HIF-1α and PHD-2 in vitro and prevents hypoxia-induced retinal neovascularization in vivo.Conclusions: AE ameliorates retinal neovascularization throuth inhibition of the HIF-1α/VEGF signaling pathway. AE may be developed as a potential drug for the prevention and treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Jianming Wu
- Laboratory of Chinese Materia Medica, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China;; Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China;; Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiao Ke
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Wei Wang
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Hongcheng Zhang
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Na Ma
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Wei Fu
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Manxi Zhao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Xiaoping Gao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Xiaofeng Hao
- Post-Doctoral Research Station, KangHong Pharmaceutical Group, Chengdu, Sichuan 610036, China
| | - Zhirong Zhang
- Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
37
|
D'Alessandro A, Nemkov T, Sun K, Liu H, Song A, Monte AA, Subudhi AW, Lovering AT, Dvorkin D, Julian CG, Kevil CG, Kolluru GK, Shiva S, Gladwin MT, Xia Y, Hansen KC, Roach RC. AltitudeOmics: Red Blood Cell Metabolic Adaptation to High Altitude Hypoxia. J Proteome Res 2016; 15:3883-3895. [PMID: 27646145 DOI: 10.1021/acs.jproteome.6b00733] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Red blood cells (RBCs) are key players in systemic oxygen transport. RBCs respond to in vitro hypoxia through the so-called oxygen-dependent metabolic regulation, which involves the competitive binding of deoxyhemoglobin and glycolytic enzymes to the N-terminal cytosolic domain of band 3. This mechanism promotes the accumulation of 2,3-DPG, stabilizing the deoxygenated state of hemoglobin, and cytosol acidification, triggering oxygen off-loading through the Bohr effect. Despite in vitro studies, in vivo adaptations to hypoxia have not yet been completely elucidated. Within the framework of the AltitudeOmics study, erythrocytes were collected from 21 healthy volunteers at sea level, after exposure to high altitude (5260 m) for 1, 7, and 16 days, and following reascent after 7 days at 1525 m. UHPLC-MS metabolomics results were correlated to physiological and athletic performance parameters. Immediate metabolic adaptations were noted as early as a few hours from ascending to >5000 m, and maintained for 16 days at high altitude. Consistent with the mechanisms elucidated in vitro, hypoxia promoted glycolysis and deregulated the pentose phosphate pathway, as well purine catabolism, glutathione homeostasis, arginine/nitric oxide, and sulfur/H2S metabolism. Metabolic adaptations were preserved 1 week after descent, consistently with improved physical performances in comparison to the first ascendance, suggesting a mechanism of metabolic memory.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver , Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver , Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Kaiqi Sun
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston , Houston, Texas, United States
| | - Hong Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston , Houston, Texas, United States
| | - Anren Song
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston , Houston, Texas, United States
| | - Andrew A Monte
- Altitude Research Center, Department of Emergency Medicine, University of Colorado , Anschutz Medical Campus, Aurora, Colorado, United States
| | - Andrew W Subudhi
- Altitude Research Center, Department of Emergency Medicine, University of Colorado , Anschutz Medical Campus, Aurora, Colorado, United States.,Department of Biology, University of Colorado Colorado Springs , Colorado Springs, Colorado, United States
| | - Andrew T Lovering
- Department of Human Physiology, University of Oregon , Eugene, Oregon, United States
| | - Daniel Dvorkin
- Altitude Research Center, Department of Emergency Medicine, University of Colorado , Anschutz Medical Campus, Aurora, Colorado, United States
| | - Colleen G Julian
- Altitude Research Center, Department of Emergency Medicine, University of Colorado , Anschutz Medical Campus, Aurora, Colorado, United States
| | - Christopher G Kevil
- Department of Pathology, Centre for Cardiovascular Diseases and Sciences, LSU Health , Shreveport, Louisiana, United States
| | - Gopi K Kolluru
- Department of Pathology, Centre for Cardiovascular Diseases and Sciences, LSU Health , Shreveport, Louisiana, United States
| | - Sruti Shiva
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania, United States
| | - Mark T Gladwin
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania, United States
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston , Houston, Texas, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver , Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robert C Roach
- Altitude Research Center, Department of Emergency Medicine, University of Colorado , Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
38
|
Wu J, Ke X, Ma N, Wang W, Fu W, Zhang H, Zhao M, Gao X, Hao X, Zhang Z. Formononetin, an active compound of Astragalus membranaceus (Fisch) Bunge, inhibits hypoxia-induced retinal neovascularization via the HIF-1α/VEGF signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3071-3081. [PMID: 27729769 PMCID: PMC5042190 DOI: 10.2147/dddt.s114022] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND It has been reported that formononetin (FMN), one of the main ingredients from famous traditional Chinese medicine "Huang-qi" (Astragalus membranaceus [Fisch] Bunge) for Qi-tonifying, exhibits the effects of immunomodulation and tumor growth inhibition via antiangiogenesis. Furthermore, A. membranaceus may alleviate the retinal neovascularization (NV) of diabetic retinopathy. However, the information of FMN on retinal NV is limited so far. In the present study, we investigated the effects of FMN on the hypoxia-induced retinal NV and the possible related mechanisms. MATERIALS AND METHODS The VEGF secretion model of acute retinal pigment epithelial-19 (ARPE-19) cells under chemical hypoxia was established by the exposure of cells to 150 μM CoCl2 and then cells were treated with 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1, a potent HIF-1α inhibitor, 1.0 μg/mL) or different concentrations of FMN (0.2 μg/mL, 1.0 μg/mL, and 5.0 μg/mL). The supernatants of cells were collected 48 hours later to measure the VEGF concentrations, following the manufacturer's instruction. The mRNA expressions of VEGF, HIF-1α, PHD-2, and β-actin were analyzed by quantitative reverse transcription polymerase chain reaction, and the protein expressions of HIF-1α and PHD-2 were determined by Western blot analysis. Furthermore, the rats with retinopathy were treated by intraperitoneal administration of conbercept injection (1.0 mg/kg) or FMN (5.0 mg/kg and 10.0 mg/kg) in an 80% oxygen atmosphere. The retinal avascular areas were assessed through visualization of the retinal vasculature by adenosine diphosphatase staining and hematoxylin and eosin staining. RESULTS FMN can indeed inhibit the VEGF secretion of ARPE-19 cells under hypoxia, downregulate the mRNA expression of VEGFA and PHD-2, and decrease the protein expression of VEGF, HIF-1α, and PHD-2 in vitro. Furthermore, FMN can prevent hypoxia-induced retinal NV in vivo. CONCLUSION FMN can ameliorate retinal NV via the HIF-1α/VEGF signaling pathway, and it may become a potential drug for the prevention and treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Jianming Wu
- Laboratory of Chinese Materia Medica, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou; Post-Doctoral Research Station, Kanghong Pharmaceutical Group; Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xiao Ke
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Na Ma
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Wei Wang
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Wei Fu
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Hongcheng Zhang
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Manxi Zhao
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Xiaoping Gao
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Xiaofeng Hao
- Post-Doctoral Research Station, Kanghong Pharmaceutical Group
| | - Zhirong Zhang
- Post-Doctoral Mobile Station, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
39
|
Hu L, Wang J, Zhu H, Wu X, Zhou L, Song Y, Zhu S, Hao M, Liu C, Fan Y, Wang Y, Li Q. Ischemic postconditioning protects the heart against ischemia-reperfusion injury via neuronal nitric oxide synthase in the sarcoplasmic reticulum and mitochondria. Cell Death Dis 2016; 7:e2222. [PMID: 27171264 PMCID: PMC4917647 DOI: 10.1038/cddis.2016.108] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 12/17/2022]
Abstract
As a result of its spatial confinement in cardiomyocytes, neuronal nitric oxide synthase (nNOS) is thought to regulate mitochondrial and sarcoplasmic reticulum (SR) function by maintaining nitroso-redox balance and Ca2+ cycling. Thus, we hypothesize that ischemic postconditioning (IPostC) protects hearts against ischemic/reperfusion (I/R) injury through an nNOS-mediated pathway. Isolated mouse hearts were subjected to I/R injury in a Langendorff apparatus, H9C2 cells and primary neonatal rat cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) in vitro. IPostC, compared with I/R, decreased infarct size and improved cardiac function, and the selective nNOS inhibitors abolished these effects. IPostC recovered nNOS activity and arginase expression. IPostC also increased AMP kinase (AMPK) phosphorylation and alleviated oxidative stress, and nNOS and AMPK inhibition abolished these effects. IPostC increased nitrotyrosine production in the cytosol but decreased it in mitochondria. Enhanced phospholamban (PLB) phosphorylation, normalized SR function and decreased Ca2+ overload were observed following the recovery of nNOS activity, and nNOS inhibition abolished these effects. Similar effects of IPostC were demonstrated in cardiomyocytes in vitro. IPostC decreased oxidative stress partially by regulating uncoupled nNOS and the nNOS/AMPK/peroxisome proliferator-activated receptor gamma coactivator 1 alpha/superoxide dismutase axis, and improved SR function through increasing SR Ca2+ load. These results suggest that IPostC protected hearts against I/R injury via an nNOS-mediated pathway.
Collapse
Affiliation(s)
- L Hu
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - J Wang
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - H Zhu
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - X Wu
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - L Zhou
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Y Song
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - S Zhu
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - M Hao
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - C Liu
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Y Fan
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Y Wang
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Q Li
- Department of Pharmacology, Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Krause BJ, Del Rio R, Moya EA, Marquez-Gutierrez M, Casanello P, Iturriaga R. Arginase-endothelial nitric oxide synthase imbalance contributes to endothelial dysfunction during chronic intermittent hypoxia. J Hypertens 2016; 33:515-24; discussion 524. [PMID: 25629363 DOI: 10.1097/hjh.0000000000000453] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Chronic intermittent hypoxia (CIH), the main feature of obstructive sleep apnoea, is associated with impaired vascular function despite unaltered response to nitric oxide donors. This study addressed whether arginase contributes to the endothelial dysfunction in CIH rats. METHODS Adult male Sprague-Dawley rats were exposed for 21 days to CIH (5% oxygen, 12 times/h, 8 h/day). The internal carotid arteries were isolated to study endothelial nitric oxide synthase (eNOS) and arginase-1 levels by western blot and immunohistochemistry, and their vasoactive responses using wire myography. Relaxation to sodium nitroprusside (SNP; nitric oxide donor) in the presence or absence of soluble guanylyl cyclase inhibitor, and acetylcholine with and without a NOS inhibitor [N(G)-nitro-L-arginine (L-NA)] and the arginase inhibitor BEC were determined. RESULTS Arteries from the CIH rats presented higher active contraction induced by KCl (3.5 ± 0.4 vs. 2.3 ± 0.2 N/m2), augmented media-to-lumen ratio (∼40%), decreased relaxation to acetylcholine (12.8 ± 1.5 vs. 30.5 ± 4.6%) and increased sensitivity to SNP (pD2 7.3 ± 0.1 vs. 6.7 ± 0.1). Arginase inhibition reversed the impaired acetylcholine-induced relaxation in CIH arteries (49.5 ± 7.4%), an effect completely blocked by L-NA. In the carotid arteries, arginase-1 protein level was increased, whereas eNOS levels decreased in the CIH arteries. CONCLUSION The current results suggest that endothelial dysfunction in CIH-induced hypertension may result from imbalanced arginase-1 to eNOS expression, vascular remodelling and increased contractile capacity, rather than decreased vascular response to nitric oxide.
Collapse
Affiliation(s)
- Bernardo J Krause
- aDivision of Obstetrics and Gynaecology bDivision of Paediatrics, Faculty of Medicine, School of Medicine cLaboratory of Neurobiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile dLaboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
41
|
Nara A, Nagai H, Shintani-Ishida K, Ogura S, Shimosawa T, Kuwahira I, Shirai M, Yoshida KI. Pulmonary arterial hypertension in rats due to age-related arginase activation in intermittent hypoxia. Am J Respir Cell Mol Biol 2015; 53:184-92. [PMID: 25490411 DOI: 10.1165/rcmb.2014-0163oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is prevalent in patients with obstructive sleep apnea syndrome (OSAS). Aging induces arginase activation and reduces nitric oxide (NO) production in the arteries. Intermittent hypoxia (IH), conferred by cycles of brief hypoxia and normoxia, contributes to OSAS pathogenesis. Here, we studied the role of arginase and aging in the pathogenesis of PAH in adult (9-mo-old) and young (2-mo-old) male Sprague-Dawley rats subjected to IH or normoxia for 4 weeks and analyzed them with a pressure-volume catheter inserted into the right ventricle (RV) and by pulsed Doppler echocardiography. Western blot analysis was conducted on arginase, NO synthase isoforms, and nitrotyrosine. IH induced PAH, as shown by increased RV systolic pressure and RV hypertrophy, in adult rats but not in young rats. IH increased expression levels of arginase I and II proteins in the adult rats. IH also increased arginase I expression in the pulmonary artery endothelium and arginase II in the pulmonary artery adventitia. Furthermore, IH reduced pulmonary levels of nitrate and nitrite but increased nitrotyrosine levels in adult rats. An arginase inhibitor (N(ω)-hydroxy-nor-1-arginine) prevented IH-induced PAH and normalized nitrite and nitrate levels in adult rats. IH induced arginase up-regulation and PAH in adult rats, but not in young rats, through reduced NO production. Our findings suggest that arginase inhibition prevents or reverses PAH.
Collapse
Affiliation(s)
- Akina Nara
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| | - Hisashi Nagai
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| | | | - Sayoko Ogura
- 1 Department of Forensic Medicine, Graduate School of Medicine and.,2 Division of Laboratory Medicine, Department of Pathology and Microbiology, Faculty of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuo Shimosawa
- 3 Department of Clinical Laboratory, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Ichiro Kuwahira
- 4 Department of Respiratory Medicine, Tokai University School of Medicine, Kanagawa, Japan; and
| | - Mikiyasu Shirai
- 5 Department of Cardiac Physiology, National Cerebral & Cardiovascular Center, Osaka, Japan
| | - Ken-ichi Yoshida
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| |
Collapse
|
42
|
Kholdani C, Fares WH, Mohsenin V. Pulmonary hypertension in obstructive sleep apnea: is it clinically significant? A critical analysis of the association and pathophysiology. Pulm Circ 2015; 5:220-7. [PMID: 26064448 DOI: 10.1086/679995] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 09/02/2014] [Indexed: 12/15/2022] Open
Abstract
The development of pulmonary hypertension is a poor prognostic sign in patients with obstructive sleep apnea (OSA) and affects both mortality and quality of life. Although pulmonary hypertension in OSA is traditionally viewed as a result of apneas and intermittent hypoxia during sleep, recent studies indicate that neither of these factors correlates very well with pulmonary artery pressure. Human data show that pulmonary hypertension in the setting of OSA is, in large part, due to left heart dysfunction with either preserved or diminished ejection fraction. Longstanding increased left heart filling pressures eventually lead to pulmonary venous hypertension. The combination of hypoxic pulmonary vasoconstriction and pulmonary venous hypertension with abnormal production of mediators will result in vascular cell proliferation and aberrant vascular remodeling leading to pulmonary hypertension. These changes are in many ways similar to those seen in other forms of pulmonary hypertension and suggest shared mechanisms. The majority of patients with OSA do not receive a diagnosis and are undertreated. Appreciating the high prevalence and understanding the mechanisms of pulmonary hypertension in OSA would lead to better recognition and management of the condition.
Collapse
Affiliation(s)
- Cyrus Kholdani
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Wassim H Fares
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Vahid Mohsenin
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
43
|
Henno P, Maurey C, Le Pimpec-Barthes F, Devillier P, Delclaux C, Israël-Biet D. Is arginase a potential drug target in tobacco-induced pulmonary endothelial dysfunction? Respir Res 2015; 16:46. [PMID: 25889611 PMCID: PMC4391310 DOI: 10.1186/s12931-015-0196-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/25/2015] [Indexed: 11/29/2022] Open
Abstract
Background Tobacco-induced pulmonary vascular disease is partly driven by endothelial dysfunction. The bioavailability of the potent vasodilator nitric oxide (NO) depends on competition between NO synthase-3 (NOS3) and arginases for their common substrate (L-arginine). We tested the hypothesis whereby tobacco smoking impairs pulmonary endothelial function via upregulation of the arginase pathway. Methods Endothelium-dependent vasodilation in response to acetylcholine (Ach) was compared ex vivo for pulmonary vascular rings from 29 smokers and 10 never-smokers. The results were expressed as a percentage of the contraction with phenylephrine. We tested the effects of L-arginine supplementation, arginase inhibition (by N(omega)-hydroxy-nor-l-arginine, NorNOHA) and NOS3 induction (by genistein) on vasodilation. Protein levels of NOS3 and arginases I and II in the pulmonary arteries were quantified by Western blotting. Results Overall, vasodilation was impaired in smokers (relative to controls; p < 0.01). Eleven of the 29 smokers (the ED+ subgroup) displayed endothelial dysfunction (defined as the absence of a relaxant response to Ach), whereas 18 (the ED− subgroup) had normal vasodilation. The mean responses to 10−4 M Ach were −23 ± 10% and 31 ± 4% in the ED+ and ED− subgroups, respectively (p < 0.01). Supplementation with L- arginine improved endothelial function in the ED+ subgroup (−4 ± 10% vs. -32 ± 10% in the presence and absence of L- arginine, respectively; p = 0.006), as did arginase inhibition (18 ± 9% vs. -1 ± 9%, respectively; p = 0.0002). Arginase I protein was overexpressed in ED+ samples, whereas ED+ and ED− samples did not differ significantly in terms of NOS3 expression. Treatment with genistein did not significantly improve endothelial function in ED+ samples. Conclusion Overexpression and elevated activity of arginase I are involved in tobacco-induced pulmonary endothelial dysfunction.
Collapse
Affiliation(s)
- Priscilla Henno
- Sorbonne Universités, UPMC Université Paris 06, Paris, France. .,Département Physiologie-Algologie-Somnologie, Unité Fonctionnelle de Somnologie et Fonction Respiratoire, AP-HP, Hôpital Saint Antoine, 75012, Paris, France. .,Laboratoire de Pharmacologie Respiratoire UPRES EA 220, Hôpital Foch, 92150, Suresnes, France.
| | - Christelle Maurey
- Ecole Nationale Vétérinaire d'Alfort, Unité de Médecine, Université Paris-Est, 94700, Maisons-Alfort, France.
| | - Françoise Le Pimpec-Barthes
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Chirurgie Thoracique, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| | - Philippe Devillier
- Laboratoire de Pharmacologie Respiratoire UPRES EA 220, Hôpital Foch, 92150, Suresnes, France. .,Université Versailles Saint-Quentin en Yvelines, UFR Sciences de la Santé Simone Veil, Montigny le Bretonneux, France.
| | - Christophe Delclaux
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Physiologie, Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| | - Dominique Israël-Biet
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Pneumologie, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| |
Collapse
|
44
|
Kobayashi S, Yamashita T, Ohneda K, Nagano M, Kimura K, Nakai H, Poellinger L, Ohneda O. Hypoxia-inducible factor-3α promotes angiogenic activity of pulmonary endothelial cells by repressing the expression of the VE-cadherin gene. Genes Cells 2015; 20:224-41. [PMID: 25626335 DOI: 10.1111/gtc.12215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/02/2014] [Indexed: 12/18/2022]
Abstract
The variants of the hypoxia-inducible factor-3α gene HIF-3α and NEPAS are known to repress the transcriptional activities driven by HIF-1α and HIF-2α. Although NEPAS has been shown to play an important role in vascular remodeling during lung development, little is known about the roles of HIF-3α in adult lung function. Here, we examined pulmonary endothelial cells (ECs) isolated from wild-type (WT) and HIF-3α functional knockout (KO) mice. The expression levels of angiogenic factors (Flk1, Ang2 and Tie2) were significantly greater in the HIF-3α KO ECs than those in the WT ECs irrespective of oxygen tension. However, the HIF-3α KO ECs showed impaired proliferative and angiogenic activities. The impaired EC function was likely due to the excess vascular endothelial (VE)-cadherin, an inhibitor of Flk1/PI3 kinase/Akt signaling, as treatment of the cells to a neutralizing antibody partly restored the phenotype of the HIF-3α KO ECs. Importantly, we found that the mRNA levels of HIF-2α and Ets-1 were significantly increased by HIF-3α ablation. Given that both factors are known to activate the VE-cadherin gene, the transcriptional repression of these factors by HIF-3α might be important for silencing the irrelevant expression of the VE-cadherin gene. Collectively, these data show novel and unique roles of HIF-3α for angiogenic gene regulation in pulmonary ECs.
Collapse
Affiliation(s)
- Satomi Kobayashi
- Graduate School of Comprehensive Human Sciences, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang L, Bhatta A, Toque HA, Rojas M, Yao L, Xu Z, Patel C, Caldwell RB, Caldwell RW. Arginase inhibition enhances angiogenesis in endothelial cells exposed to hypoxia. Microvasc Res 2014; 98:1-8. [PMID: 25445030 DOI: 10.1016/j.mvr.2014.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/29/2014] [Accepted: 11/02/2014] [Indexed: 10/24/2022]
Abstract
Hypoxia-induced arginase elevation plays an essential role in several vascular diseases but influence of arginase on hypoxia-mediated angiogenesis is completely unknown. In this study, in vitro network formation in bovine aortic endothelial cells (BAEC) was examined after exposure to hypoxia for 24h with or without arginase inhibition. Arginase activity, protein levels of the two arginase isoforms, eNOS, and VEGF as well as production of NO and ROS were examined to determine the involvement of arginase in hypoxia-mediated angiogenesis. Hypoxia elevated arginase activity and arginase 2 expression but reduced active p-eNOS(Ser1177) and NO levels in BAEC. In addition, both VEGF protein levels and endothelial elongation and network formation were reduced with continued hypoxia, whereas ROS levels increased and NO levels decreased. Arginase inhibition limited ROS, restored NO formation and VEGF expression, and prevented the reduction of angiogenesis. These results suggest a fundamental role of arginase activity in regulating angiogenic function.
Collapse
Affiliation(s)
- Lin Wang
- Department of Plastic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Anil Bhatta
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Modesto Rojas
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Zhimin Xu
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Chintan Patel
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
46
|
Suwanpradid J, Rojas M, Behzadian MA, Caldwell RW, Caldwell RB. Arginase 2 deficiency prevents oxidative stress and limits hyperoxia-induced retinal vascular degeneration. PLoS One 2014; 9:e110604. [PMID: 25375125 PMCID: PMC4222858 DOI: 10.1371/journal.pone.0110604] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/16/2014] [Indexed: 01/02/2023] Open
Abstract
Background Hyperoxia exposure of premature infants causes obliteration of the immature retinal microvessels, leading to a condition of proliferative vitreoretinal neovascularization termed retinopathy of prematurity (ROP). Previous work has demonstrated that the hyperoxia-induced vascular injury is mediated by dysfunction of endothelial nitric oxide synthase resulting in peroxynitrite formation. This study was undertaken to determine the involvement of the ureahydrolase enzyme arginase in this pathology. Methods and Findings Studies were performed using hyperoxia-treated bovine retinal endothelial cells (BRE) and mice with oxygen-induced retinopathy (OIR) as experimental models of ROP. Treatment with the specific arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) prevented hyperoxia-induced apoptosis of BRE cells and reduced vaso-obliteration in the OIR model. Furthermore, deletion of the arginase 2 gene protected against hyperoxia-induced vaso-obliteration, enhanced physiological vascular repair, and reduced retinal neovascularization in the OIR model. Additional deletion of one copy of arginase 1 did not improve the vascular pathology. Analyses of peroxynitrite by quantitation of its biomarker nitrotyrosine, superoxide by dihydroethidium imaging and NO formation by diaminofluoroscein imaging showed that the protective actions of arginase 2 deletion were associated with blockade of superoxide and peroxynitrite formation and normalization of NOS activity. Conclusions Our data demonstrate the involvement of arginase activity and arginase 2 expression in hyperoxia-induced vascular injury. Arginase 2 deletion prevents hyperoxia-induced retinal vascular injury by preventing NOS uncoupling resulting in decreased reactive oxygen species formation and increased nitric oxide bioavailability.
Collapse
Affiliation(s)
- Jutamas Suwanpradid
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
| | - Modesto Rojas
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology and Toxicology, Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - M. Ali Behzadian
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - R. William Caldwell
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology and Toxicology, Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Fike CD, Summar M, Aschner JL. L-citrulline provides a novel strategy for treating chronic pulmonary hypertension in newborn infants. Acta Paediatr 2014; 103:1019-26. [PMID: 24862864 DOI: 10.1111/apa.12707] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/23/2014] [Accepted: 05/23/2014] [Indexed: 02/05/2023]
Abstract
UNLABELLED Effective therapies are urgently needed for infants with forms of pulmonary hypertension that develop or persist beyond the first week of life. The L-arginine nitric oxide (NO) precursor, L-citrulline, improves NO signalling and ameliorates pulmonary hypertension in newborn animals. In vitro studies demonstrate that manipulating L-citrulline transport alters NO production. CONCLUSION Strategies that increase the supply and transport of L-citrulline merit pursuit as novel approaches to managing infants with chronic, progressive pulmonary hypertension.
Collapse
Affiliation(s)
- Candice D. Fike
- Department of Pediatrics; Vanderbilt University Medical Center; Nashville TN USA
- Monroe Carell Jr. Children's Hospital at Vanderbilt; Nashville TN USA
| | - Marshall Summar
- Division of Genetics and Metabolism; Children's National Medical Center; Washington DC USA
| | - Judy L. Aschner
- Department of Pediatrics; Albert Einstein College of Medicine and the Children's Hospital at Montefiore; New York NY USA
| |
Collapse
|
48
|
Toya T, Hakuno D, Shiraishi Y, Kujiraoka T, Adachi T. Arginase inhibition augments nitric oxide production and facilitates left ventricular systolic function in doxorubicin-induced cardiomyopathy in mice. Physiol Rep 2014; 2:2/9/e12130. [PMID: 25263201 PMCID: PMC4270236 DOI: 10.14814/phy2.12130] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A metabolizing enzyme arginase can decrease nitric oxide (NO) production by competing with NO synthase for arginine as a substrate, but its pathophysiological role in heart failure remains unknown. We aimed to investigate the effect of pharmacological inhibition of arginase on left ventricular function in doxorubicin‐induced cardiomyopathy in mice. Doxorubicin administration for 5 weeks significantly increased protein expression levels or activity of arginase in the lungs and liver, and caused moderate increase in arginase 2 expression in the aorta. In the lungs, accumulated interstitial cells strongly expressed both arginase 1 and arginase 2 by doxorubicin administration. Echocardiography revealed that administration of a potent, reversible arginase inhibitor N‐omega‐hydroxy‐nor‐l‐arginine completely reversed doxorubicin‐induced decrease in the ejection fraction, in parallel with expression levels of BNP mRNA, without affecting apoptosis, hypertrophy, fibrosis, or macrophage infiltration in the left ventricle. Arginase inhibition reversibly lowered systolic blood pressure, and importantly, it recovered doxorubicin‐induced decline in NO concentration in the serum, lungs, and aorta. Furthermore, arginase inhibition stimulated NO secretion from aortic endothelial cells and peritoneal macrophages in vitro. In conclusion, pharmacological inhibition of arginase augmented NO concentration in the serum, lungs, and aorta, promoted NO‐mediated decrease in afterload for left ventricle, and facilitated left ventricular systolic function in doxorubicin‐induced cardiomyopathy in mice. e12130 This figure shows that administration of an arginase inhibitor nor‐ NOHA facilitates LV systolic function in murine Dox‐induced cardiomyopathy.
Collapse
Affiliation(s)
- Takumi Toya
- Division of Cardiology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Daihiko Hakuno
- Division of Cardiology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yasunaga Shiraishi
- Division of Cardiology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Takehiko Kujiraoka
- Division of Cardiology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Takeshi Adachi
- Division of Cardiology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
49
|
Huang S, Chen P, Shui X, He Y, Wang H, Zheng J, Zhang L, Li J, Xue Y, Chen C, Lei W. Baicalin attenuates transforming growth factor-β1-induced human pulmonary artery smooth muscle cell proliferation and phenotypic switch by inhibiting hypoxia inducible factor-1α and aryl hydrocarbon receptor expression. J Pharm Pharmacol 2014; 66:1469-77. [PMID: 24835111 DOI: 10.1111/jphp.12273] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/13/2014] [Indexed: 12/20/2022]
Abstract
Abstract
Objectives
Baicalin, a natural flavone, has antithrombotic, antihyperlipidemic and antiinflammortory activity. It can also inhibit cancer cell proliferation and reduce brain cell apoptosis. This study aimed to elucidate the effect of baicalin on the excessive proliferation of human pulmonary arterial smooth muscle cells (HPASMCs) induced by transforming growth factor-β1 (TGF-β1) and to investigate the roles of hypoxia inducible factor-1α (HIF-1α) and aryl hydrocarbon receptor (AhR) in mediating this TGF-β1-induced excessive proliferation of HPASMCs.
Methods
TGF-β1-induced proliferation of HPASMCs was assayed using the CCK8 method. The cellular phenotype was identified by immunocytochemical staining. Expression of HIF-1α and AhR mRNA was determined by real-time quantitative PCR.
Key findings
TGF-β1 promoted significantly HPASMC proliferation (P < 0.05) and induced a phenotypic switch from the contractile to synthetic type. Baicalin inhibited this TGF-β1-induced phenotypic switch and consequently the excessive growth of HPASMCs in a time-dependent and dose-dependent manner (P < 0.05). Furthermore, baicalin attenuated the abnormal proliferation of HPASMCs through suppression of the HIF-1α and AhR pathways.
Conclusions
Our study shows that baicalin has the potential to be used as a novel drug in the treatment of pulmonary arterial hypertension pathology by antagonizing HIF-1α and AhR expression and subsequently decreasing HPASMC proliferation and the phenotypic switch.
Collapse
Affiliation(s)
- Shian Huang
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Puwen Chen
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Xiaorong Shui
- Vascular Surgery Laboratory, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Yuan He
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Heyong Wang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Zheng
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI, USA
| | - Liangqing Zhang
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Jianwen Li
- Vascular Surgery Laboratory, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Yiqiang Xue
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Can Chen
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Wei Lei
- Laboratory of Cardiovascular Remodeling and Pharmaceutical Biotechnology, Department of Cardiovascular, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| |
Collapse
|
50
|
Peroxynitrite and peroxiredoxin in the pathogenesis of experimental amebic liver abscess. BIOMED RESEARCH INTERNATIONAL 2014; 2014:324230. [PMID: 24822193 PMCID: PMC4009108 DOI: 10.1155/2014/324230] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
The molecular mechanisms by which Entamoeba histolytica causes amebic liver abscess (ALA) are still not fully understood. Amebic mechanisms of adherence and cytotoxic activity are pivotal for amebic survival but apparently do not directly cause liver abscess. Abundant evidence indicates that chronic inflammation (resulting from an inadequate immune response) is probably the main cause of ALA. Reports referring to inflammatory mechanisms of liver damage mention a repertoire of toxic molecules by the immune response (especially nitric oxide and reactive oxygen intermediates) and cytotoxic substances released by neutrophils and macrophages after being lysed by amoebas (e.g., defensins, complement, and proteases). Nevertheless, recent evidence downplays these mechanisms in abscess formation and emphasizes the importance of peroxynitrite (ONOO−). It seems that the defense mechanism of amoebas against ONOO−, namely, the amebic thioredoxin system (including peroxiredoxin), is superior to that of mammals. The aim of the present text is to define the importance of ONOO− as the main agent of liver abscess formation during amebic invasion, and to explain the superior capacity of amoebas to defend themselves against this toxic agent through the peroxiredoxin and thioredoxin system.
Collapse
|