1
|
Yeudall S, Upchurch CM, Leitinger N. The clinical relevance of heme detoxification by the macrophage heme oxygenase system. Front Immunol 2024; 15:1379967. [PMID: 38585264 PMCID: PMC10995405 DOI: 10.3389/fimmu.2024.1379967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Heme degradation by the heme oxygenase (HMOX) family of enzymes is critical for maintaining homeostasis and limiting heme-induced tissue damage. Macrophages express HMOX1 and 2 and are critical sites of heme degradation in healthy and diseased states. Here we review the functions of the macrophage heme oxygenase system and its clinical relevance in discrete groups of pathologies where heme has been demonstrated to play a driving role. HMOX1 function in macrophages is essential for limiting oxidative tissue damage in both acute and chronic hemolytic disorders. By degrading pro-inflammatory heme and releasing anti-inflammatory molecules such as carbon monoxide, HMOX1 fine-tunes the acute inflammatory response with consequences for disorders of hyperinflammation such as sepsis. We then discuss divergent beneficial and pathological roles for HMOX1 in disorders such as atherosclerosis and metabolic syndrome, where activation of the HMOX system sits at the crossroads of chronic low-grade inflammation and oxidative stress. Finally, we highlight the emerging role for HMOX1 in regulating macrophage cell death via the iron- and oxidation-dependent form of cell death, ferroptosis. In summary, the importance of heme clearance by macrophages is an active area of investigation with relevance for therapeutic intervention in a diverse array of human diseases.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Clint M. Upchurch
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
2
|
Luo JY, Cheng CK, He L, Pu Y, Zhang Y, Lin X, Xu A, Lau CW, Tian XY, Ma RCW, Jo H, Huang Y. Endothelial UCP2 Is a Mechanosensitive Suppressor of Atherosclerosis. Circ Res 2022; 131:424-441. [PMID: 35899624 PMCID: PMC9390236 DOI: 10.1161/circresaha.122.321187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Inflamed endothelial cells (ECs) trigger atherogenesis, especially at arterial regions experiencing disturbed blood flow. UCP2 (Uncoupling protein 2), a key mitochondrial antioxidant protein, improves endothelium-dependent relaxation in obese mice. However, whether UCP2 can be regulated by shear flow is unknown, and the role of endothelial UCP2 in regulating inflammation and atherosclerosis remains unclear. This study aims to investigate the mechanoregulation of UCP2 expression in ECs and the effect of UCP2 on endothelial inflammation and atherogenesis. METHODS In vitro shear stress simulation system was used to investigate the regulation of UCP2 expression by shear flow. EC-specific Ucp2 knockout mice were used to investigate the role of UCP2 in flow-associated atherosclerosis. RESULTS Shear stress experiments showed that KLF2 (Krüppel-like factor 2) mediates fluid shear stress-dependent regulation of UCP2 expression in human aortic and human umbilical vein ECs. Unidirectional shear stress, statins, and resveratrol upregulate whereas oscillatory shear stress and proinflammatory stimuli inhibit UCP2 expression through altered KLF2 expression. KLF2 directly binds to UCP2 promoter to upregulate its transcription in human umbilical vein ECs. UCP2 knockdown induced expression of genes involved in proinflammatory and profibrotic signaling, resulting in a proatherogenic endothelial phenotype. EC-specific Ucp2 deletion promotes atherogenesis and collagen production. Additionally, we found endothelial Ucp2 deficiency aggravates whereas adeno-associated virus-mediated EC-Ucp2 overexpression inhibits carotid atherosclerotic plaque formation in disturbed flow-enhanced atherosclerosis mouse model. RNA-sequencing analysis revealed FoxO1 (forkhead box protein O1) as the major proinflammatory transcriptional regulator activated by UCP2 knockdown, and FoxO1 inhibition reduced vascular inflammation and disturbed flow-enhanced atherosclerosis. We showed further that UCP2 level is critical for phosphorylation of AMPK (AMP-activated protein kinase), which is required for UCP2-induced inhibition of FoxO1. CONCLUSIONS Altogether, our studies uncover that UCP2 is novel mechanosensitive gene under the control of fluid shear stress and KLF2 in ECs. UCP2 expression is critical for endothelial proinflammatory response and atherogenesis. Therapeutic strategies enhancing UCP2 level may have therapeutic potential against atherosclerosis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China (J.-Y.L.)
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Lei He
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yujie Pu
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China (Y.Z.)
| | - Xiao Lin
- School of Life Sciences (X.L.), Chinese University of Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, China (A.X.)
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Xiao Yu Tian
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics (R.C.W.M.), Chinese University of Hong Kong, China
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (H.J.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| |
Collapse
|
3
|
Ameka M, Hasty AH. Paying the Iron Price: Liver Iron Homeostasis and Metabolic Disease. Compr Physiol 2022; 12:3641-3663. [PMID: 35766833 PMCID: PMC10155403 DOI: 10.1002/cphy.c210039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Iron is an essential metal element whose bioavailability is tightly regulated. Under normal conditions, systemic and cellular iron homeostases are synchronized for optimal function, based on the needs of each system. During metabolic dysfunction, this synchrony is lost, and markers of systemic iron homeostasis are no longer coupled to the iron status of key metabolic organs such as the liver and adipose tissue. The effects of dysmetabolic iron overload syndrome in the liver have been tied to hepatic insulin resistance, nonalcoholic fatty liver disease, and nonalcoholic steatohepatitis. While the existence of a relationship between iron dysregulation and metabolic dysfunction has long been acknowledged, identifying correlative relationships is complicated by the prognostic reliance on systemic measures of iron homeostasis. What is lacking and perhaps more informative is an understanding of how cellular iron homeostasis changes with metabolic dysfunction. This article explores bidirectional relationships between different proteins involved in iron homeostasis and metabolic dysfunction in the liver. © 2022 American Physiological Society. Compr Physiol 12:3641-3663, 2022.
Collapse
Affiliation(s)
- Magdalene Ameka
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Yao L, Hao Y, Wen G, Xiao Q, Wu P, Wang J, Liu J. Induction of Heme Oxygenase-1 Modifies the Systemic Immunity and Reduces Atherosclerotic Lesion Development in ApoE Deficient Mice. Front Pharmacol 2022; 13:809469. [PMID: 35281895 PMCID: PMC8908104 DOI: 10.3389/fphar.2022.809469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) has been reported to protect against oxidation and inflammation in atherosclerosis. It remains unclear how the immune system participates in the cytoprotective function of HO-1 in the context of atherosclerosis. In this study, we attempted to investigate the potential effect of a HO-1 inducer, hemin, and a HO-1 inhibitor, Tin-protoporphyrin IX (SnPP), on the progression of atherosclerosis in ApoE deficient mice. Using mass cytometry, 15 immune cell populations and 29 T cell sub-clusters in spleen and peripheral blood were thoroughly analyzed after hemin or SnPP treatment. SnPP elevated risk factors of atherosclerosis, whereas hemin reduced them. In-depth analysis showed that hemin significantly modified the immune system in both spleen and peripheral blood. Hemin increased dendritic (DC) and myeloid-derived suppressor cells (MDSCs), but decreased natural killer (NK) cells. An opposite effect was observed with SnPP treatment in terms of NK cells. NK cells and MDSCs were positively and negatively correlated with total cholesterol and low-density lipoprotein, respectively. Moreover, the T cell profiles were significantly reshaped by hemin, whereas only minor changes were observed with SnPP. Several hemin-modulated T cell clusters associated with atherosclerosis were also identified. In summary, we have unraveled an important regulatory role for HO-1 pathway in immune cell regulation and atherosclerosis. Our finding suggests that modulating HO-1 signaling represents a potential therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
- Leyi Yao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Institute of Digestive Disease of Guangzhou Medical University, Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yali Hao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Guanmei Wen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qingzhong Xiao
- Clinical Pharmacology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Penglong Wu
- Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinheng Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Garcia C, Blesso CN. Antioxidant properties of anthocyanins and their mechanism of action in atherosclerosis. Free Radic Biol Med 2021; 172:152-166. [PMID: 34087429 DOI: 10.1016/j.freeradbiomed.2021.05.040] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/14/2021] [Accepted: 05/29/2021] [Indexed: 12/20/2022]
Abstract
Atherosclerosis develops due to lipid accumulation in the arterial wall and sclerosis as result of increased hyperlipidemia, oxidative stress, lipid oxidation, and protein oxidation. However, improving antioxidant status through diet may prevent the progression of atherosclerotic cardiovascular disease. It is believed that polyphenol-rich plants contribute to the inverse relationship between fruit and vegetable intake and chronic disease. Anthocyanins are flavonoid polyphenols with antioxidant properties that have been associated with reduced risk of cardiovascular disease. The consumption of anthocyanins increases total antioxidant capacity, antioxidant defense enzymes, and HDL antioxidant properties by several measures in preclinical and clinical populations. Anthocyanins appear to impart antioxidant actions via direct antioxidant properties, as well as indirectly via inducing intracellular Nrf2 activation and antioxidant gene expression. These actions counter oxidative stress and inflammatory signaling in cells present in atherosclerotic plaques, including macrophages and endothelial cells. Overall, anthocyanins may protect against atherosclerosis and cardiovascular disease through their effects on cellular antioxidant status, oxidative stress, and inflammation; however, their underlying mechanisms of action appear to be complex and require further elucidation.
Collapse
Affiliation(s)
- Chelsea Garcia
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06269, United States
| | - Christopher N Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, 06269, United States.
| |
Collapse
|
6
|
Ratajczak MZ, Adamiak M, Ratajczak J, Kucia M. Heme Oxygenase 1 (HO-1) as an Inhibitor of Trafficking of Normal and Malignant Hematopoietic Stem Cells - Clinical and Translational Implications. Stem Cell Rev Rep 2021; 17:821-828. [PMID: 33196976 PMCID: PMC8166705 DOI: 10.1007/s12015-020-10083-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/12/2022]
Abstract
Evidence indicates that bone marrow (BM)-residing hematopoietic stem/progenitor cells (HSPCs) are released into peripheral blood (PB) after administration of pro-mobilizing drugs, which induce a state of sterile inflammation in the BM microenvironment. In the reverse process, as seen after hematopoietic transplantation, intravenously injected HSPCs home and engraft into BM niches. Here again, conditioning for transplantation by myeloablative chemo- or radiotherapy induces a state of sterile inflammation that promotes HSPC seeding to BM stem cell niches. Therefore, the trafficking of HSPCs and their progeny, including granulocytes and monocytes/macrophages, is regulated by a response to pro-inflammatory stimuli. This responsiveness to inflammatory cues is also preserved after malignant transformation of hematopoietic cells. Results from our laboratory indicate that the responsiveness of hematopoietic cells to pro-inflammatory stimuli is orchestrated by Nlrp3 inflammasome. As reported, HO-1 effectively attenuates intracellular activation of Nlrp3 inflammasome as well as the pro-inflammatory effects of several humoral mediators, including complement cascade (ComC) cleavage fragments that promote migration of hematopoietic cells. Based on this finding, inhibition of HO-1 activity may become a practical strategy to enhance the mobilization and homing of normal HSPCs, and, alternatively, its activation may prevent unwanted spread and in vivo expansion of leukemic cells. Graphical Abstract.
Collapse
Affiliation(s)
- Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
7
|
Yao ME, Su MY, Huang Y, Chen W. Physiologically increased total bilirubin is associated with reduced risk of first myocardial infarction: A meta-analysis and dose-response analysis. Nutr Metab Cardiovasc Dis 2021; 31:1016-1026. [PMID: 33612380 DOI: 10.1016/j.numecd.2021.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/10/2023]
Abstract
AIM Bilirubin has potential predictive and prognostic value for myocardial infarction (MI), but the clinical evidence remains controversial. We performed this meta-analysis to systematically quantify the relationships between circulating bilirubin levels and the incidence of MI and post-MI adverse events. DATA SYNTHESIS We searched the PubMed, Cochrane Library, Embase, and Web of Science databases for ad-hoc studies, published up to October 17, 2020, recording bilirubin before MI (predictive analyses) or adverse events (prognostic analyses). Relative risks (RR) were pooled by a random-effects model. The dose-response analysis was conducted by restricted cubic splines. In patients without previous MI, increased total bilirubin (TB) reduced the risk of long-term (>3 year) first MI by 22% (95% confidence interval [CI]: 0.69-0.88, n = 4). The dose-response analysis indicated that the RR for first MI decreased by 2.7% per each 2 μmol/L increment of TB (three studies, 95% CI: 1.3%-4.1%, P < 0.001), with a cut-off value of 12.60 μmol/L for RR > 1.00. Elevated bilirubin reduced the incidence of first and recurrent MI by 36% (95% CI: 0.42-0.98, n = 7). However, after suffering MI, higher TB concentrations could not decrease the risk of recurrent MI (RR: 1.02, 95% CI: 0.67-1.55, n = 5) and increased the incidence of short-term (<1 year) post-MI major adverse cardiovascular events, all-cause mortality, and cardiovascular mortality, but not long-term (≥1 year). CONCLUSION Higher TB levels within a physiological range reduced the incidence of long-term first MI, with a cut-off value of 12.60 μmol/L.
Collapse
Affiliation(s)
- Miao-En Yao
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Road, Zhangjiang Hi-TechPark, Pudong New Area, Shanghai, 201203, China
| | - Mei-Yi Su
- Second Clinical College of Guangzhou University of Chinese Medicine, NO.232, Outer-ring East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China; Department of Cardiovascular Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, NO.111, Dade Road, Yue-Xiu District, Guangzhou, 510120, China
| | - Yi Huang
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Road, Zhangjiang Hi-TechPark, Pudong New Area, Shanghai, 201203, China
| | - Wei Chen
- Department of Critical Care Medicine, Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
8
|
Hosseini V, Mallone A, Nasrollahi F, Ostrovidov S, Nasiri R, Mahmoodi M, Haghniaz R, Baidya A, Salek MM, Darabi MA, Orive G, Shamloo A, Dokmeci MR, Ahadian S, Khademhosseini A. Healthy and diseased in vitro models of vascular systems. LAB ON A CHIP 2021; 21:641-659. [PMID: 33507199 DOI: 10.1039/d0lc00464b] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Irregular hemodynamics affects the progression of various vascular diseases, such atherosclerosis or aneurysms. Despite the extensive hemodynamics studies on animal models, the inter-species differences between humans and animals hamper the translation of such findings. Recent advances in vascular tissue engineering and the suitability of in vitro models for interim analysis have increased the use of in vitro human vascular tissue models. Although the effect of flow on endothelial cell (EC) pathophysiology and EC-flow interactions have been vastly studied in two-dimensional systems, they cannot be used to understand the effect of other micro- and macro-environmental parameters associated with vessel wall diseases. To generate an ideal in vitro model of the vascular system, essential criteria should be included: 1) the presence of smooth muscle cells or perivascular cells underneath an EC monolayer, 2) an elastic mechanical response of tissue to pulsatile flow pressure, 3) flow conditions that accurately mimic the hemodynamics of diseases, and 4) geometrical features required for pathophysiological flow. In this paper, we review currently available in vitro models that include flow dynamics and discuss studies that have tried to address the criteria mentioned above. Finally, we critically review in vitro fluidic models of atherosclerosis, aneurysm, and thrombosis.
Collapse
Affiliation(s)
- Vahid Hosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Anna Mallone
- Institute of Regenerative Medicine, University of Zurich, Zurich CH-8952, Switzerland
| | - Fatemeh Nasrollahi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Serge Ostrovidov
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and Department of Radiological Sciences, University of California-Los Angeles, CA 90095, USA
| | - Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Department of Mechanical Engineering, Sharif University of Technology, Tehran 1136511155, Iran
| | - Mahboobeh Mahmoodi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd 8915813135, Iran
| | - Reihaneh Haghniaz
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Avijit Baidya
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA
| | - M Mehdi Salek
- School of Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain and Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01007, Spain
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 1136511155, Iran
| | - Mehmet R Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, CA 90095, USA and California NanoSystems Institute and Department of Bioengineering, University of California-Los Angeles, CA 90095, USA and Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| |
Collapse
|
9
|
Khojandi N, Kuehm LM, Piening A, Donlin MJ, Hsueh EC, Schwartz TL, Farrell K, Richart JM, Geerling E, Pinto AK, George SL, Albert CJ, Ford DA, Chen X, Kline J, Teague RM. Oxidized Lipoproteins Promote Resistance to Cancer Immunotherapy Independent of Patient Obesity. Cancer Immunol Res 2021; 9:214-226. [PMID: 33303575 PMCID: PMC7864876 DOI: 10.1158/2326-6066.cir-20-0358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/07/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022]
Abstract
Antitumor immunity is impaired in obese mice. Mechanistic insight into this observation remains sparse and whether it is recapitulated in patients with cancer is unclear because clinical studies have produced conflicting and controversial findings. We addressed this by analyzing data from patients with a diverse array of cancer types. We found that survival after immunotherapy was not accurately predicted by body mass index or serum leptin concentrations. However, oxidized low-density lipoprotein (ox-LDL) in serum was identified as a suppressor of T-cell function and a driver of tumor cytoprotection mediated by heme oxygenase-1 (HO-1). Analysis of a human melanoma gene expression database showed a clear association between higher HMOX1 (HO-1) expression and reduced progression-free survival. Our in vivo experiments using mouse models of both melanoma and breast cancer revealed HO-1 as a mechanism of resistance to anti-PD1 immunotherapy but also exposed HO-1 as a vulnerability that could be exploited therapeutically using a small-molecule inhibitor. In conclusion, our clinical data have implicated serum ox-LDL as a mediator of therapeutic resistance in patients with cancer, operating as a double-edged sword that both suppressed T-cell immunity and simultaneously induced HO-1-mediated tumor cell protection. Our studies also highlight the therapeutic potential of targeting HO-1 during immunotherapy, encouraging further translational development of this combination approach.See article by Kuehm et al., p. 227.
Collapse
Affiliation(s)
- Niloufar Khojandi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Lindsey M Kuehm
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Alexander Piening
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Maureen J Donlin
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Eddy C Hsueh
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Theresa L Schwartz
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Kaitlin Farrell
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri
| | - John M Richart
- Department of Internal Medicine, Division of Hematology and Oncology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Sarah L George
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Carolyn J Albert
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Xiufen Chen
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Justin Kline
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ryan M Teague
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri.
- Alvin J. Siteman National Cancer Institute Comprehensive Cancer Center, St. Louis, Missouri
| |
Collapse
|
10
|
Shiels RG, Hewage W, Pennell EN, Vidimce J, Grant G, Pearson AG, Wagner KH, Morgan M, Bulmer AC. Biliverdin and bilirubin sulfonate inhibit monosodium urate induced sterile inflammation in the rat. Eur J Pharm Sci 2020; 155:105546. [PMID: 32927072 DOI: 10.1016/j.ejps.2020.105546] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 08/07/2020] [Accepted: 08/31/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Biliverdin, a by-product of haem catabolism, possesses potent endogenous antioxidant and anti-inflammatory properties. Bilirubin-C10-sulfonate (BRS), an active metabolite formed after enteral administration of BV in the rat, also possess antioxidant properties. Therefore, we investigated the anti-inflammatory and antioxidant activity of BV and BRS in an in vivo model of monosodium urate induced sterile inflammation. METHODS Subcutaneous air pouches were created on the dorsal flanks of Wistar rats (10-12 weeks of age). Prior to stimulation of the 6-day old pouch with monosodium urate (25 mg), groups were pre-treated with intraperitoneal BRS (27 mg/kg) and BV (27 mg/kg). Total and differential leukocyte counts were determined in pouch fluid aspirate at 1, 6, 12, 24 and 48 h after monosodium urate stimulation. Biliverdin (BV), BRS and unconjugated bilirubin (UCB) concentrations in the serum and pouch fluid were quantified using liquid chromatography-mass spectrometry. Pouch fluid cytokine concentrations (IL-1β, IL-1α, TNF-α, IL-17A, IL-12, GM-CSF, IL-33, IFN-γ, IL-18, IL-10, MCP-1, CXCL-1 and IL-6) were assessed after 6 h. In addition, 24 h protein carbonyl and chloramine concentrations were assessed in pouch fluid using ELISA and spectrophotometry, respectively. RESULTS BRS and BV significantly (p < 0.05) inhibited leukocyte (total, neutrophil and macrophage) infiltration into the pouch fluid from 6 to 48 h. For example, after 6 h neutrophil counts decreased following BRS (0.32 ± 0.11 × 106 cells mL-1) and BV (0.17 ± 0.03 × 106 cells mL-1) compared to MSU only (3.51 ± 1.07 × 106 cells mL-1). Both BV and BRS significantly (p < 0.05) reduced pouch GM-CSF (BV: 5.8 ± 1.2 pg mL-1, BRS: 6.9 ± 1.5 pg mL-1 vs MSU only: 13.0 ± 1.9 pg mL-1) and MCP-1 concentrations at 6 h (BV: 1804 ± 269 pg mL-1, BRS: 7927 ± 2668 pg mL-1 vs MSU only: 17,290 ± 4503 pg ml-1), whilst BV additionally inhibited IL-6 (4354 ± 977 pg mL-1 vs MSU only: 25,070 ± 5178 pg mL-1) and IL-18 (17.6 ± 2.0 pg mL-1 vs MSU only: 81.5 ± 19.9 pg mL-1) concentrations at 6 h (p < 0.05). Despite these differences, no change in pouch chloramine or protein carbonyl concentrations occurred at 24 h (p > 0.05). Serum BV concentrations rapidly diminished over 6 h, however, BRS was readily detected in the serum over 48 h, and in pouch fluid over 12 h. CONCLUSIONS This study is the first to elucidate anti-inflammatory activity of BRS and the efficacy of BV administration in a model of gouty inflammation. Reduced leukocyte infiltration and cytokine production in response to sterile inflammation further support the importance of these molecules in physiology and their therapeutic potential in sterile inflammation.
Collapse
Affiliation(s)
- Ryan G Shiels
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Wenu Hewage
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Evan N Pennell
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Josif Vidimce
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Gary Grant
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Andrew G Pearson
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Michael Morgan
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew C Bulmer
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
11
|
Li D, Zhao D, Du J, Dong S, Aldhamin Z, Yuan X, Li W, Du H, Zhao W, Cui L, Liu L, Fu N, Nan Y. Heme oxygenase-1 alleviated non-alcoholic fatty liver disease via suppressing ROS-dependent endoplasmic reticulum stress. Life Sci 2020; 253:117678. [PMID: 32376267 DOI: 10.1016/j.lfs.2020.117678] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022]
Abstract
AIMS The endoplasmic reticulum (ER) stress response plays a crucial role in the development of nonalcoholic steatohepatitis (NASH). Heme oxygenase-1 (HO-1) exerts beneficial effects against oxidative injury in NASH. This study is aimed to clarify whether HO-1 is an effective therapeutic strategy for NASH via regulation of ER stress. METHODS The C57BL/6J mice were fed with methionine-choline deficient (MCD) for 4 weeks and high fat-high carbohydrate-high cholesterol (HFD) diet for 16 weeks, with hemin or zinc protoporphyrin IX (ZnPP-IX), respectively. The LO-2 cells were cultured in palmitic medium, with transfected pEX-HO-1 or sh-HO-1 plasmid for 24 h. Meanwhile, thirty NASH patients and 15 health controls were enrolled. The ER ultrastructure was observed by transmission electron microscopy (TEM) and confocal microscopy. The expressions of mRNAs and proteins of HO-1, ER stress related genes were detected by real time PCR, western blot and immunohistochemical staining, respectively. RESULTS The swelled and broken rough endoplasmic reticulums were observed in MCD and HFD fed mice. The reactive hepatic expression of HO-1 was related with the increased ROS production and ER stress, companied with upregulation of GRP78, p-IRE1, PERK, ATF6. Through hemin administration, hepatocyte apoptosis was suppressed companied down-regulation of CHOP, caspase12 and up-regulation of BCL2. Conserved results were exhibited in ZnPP-IX administrated mice and HO-1 silent cells. Consistent results were observed in the NASH Patients. CONCLUSIONS HO-1 could serve as a protective factor in the progression of nutritional steatohepatitis by suppresses hepatocyte excessive ER stress and might be a potential target for therapy of nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Dandan Zhao
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Jinghua Du
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Shiming Dong
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Zaid Aldhamin
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Xiwei Yuan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Wencong Li
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Huijuan Du
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Wen Zhao
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Luyao Cui
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Lingdi Liu
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Na Fu
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, 050051 Shijiazhuang, China; Hebei Provincial Key Laboratory of Liver Fibrosis in Chronic Liver Diseases, China.
| |
Collapse
|
12
|
Karki P, Birukov KG. Oxidized Phospholipids in Healthy and Diseased Lung Endothelium. Cells 2020; 9:cells9040981. [PMID: 32326516 PMCID: PMC7226969 DOI: 10.3390/cells9040981] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating and cell membrane phospholipids undergo oxidation caused by enzymatic and non-enzymatic mechanisms. As a result, a diverse group of bioactive oxidized phospholipids generated in these conditions have both beneficial and harmful effects on the human body. Increased production of oxidized phospholipid products with deleterious effects is linked to the pathogenesis of various cardiopulmonary disorders such as atherosclerosis, thrombosis, acute lung injury (ALI), and inflammation. It has been determined that the contrasting biological effects of lipid oxidation products are governed by their structural variations. For example, full-length products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine oxidation (OxPAPC) have prominent endothelial barrier protective and anti-inflammatory activities while most of the truncated oxidized phospholipids induce vascular leak and exacerbate inflammation. The extensive studies from our group and other groups have demonstrated a strong potential of OxPAPC in mitigating a wide range of agonist-induced lung injuries and inflammation in pulmonary endothelial cell culture and rodent models of ALI. Concurrently, elevated levels of truncated oxidized phospholipids are present in aged mice lungs that potentiate the inflammatory agents-induced lung injury. On the other hand, increased levels of full length OxPAPC products accelerate ALI recovery by facilitating production of anti-inflammatory lipid mediator, lipoxin A4, and other molecules with anti-inflammatory properties. These findings suggest that OxPAPC-assisted lipid program switch may be a promising therapeutic strategy for treatment of acute inflammatory syndromes. In this review, we will summarize the vascular-protective and deleterious aspects of oxidized phospholipids and discuss their therapeutic potential including engineering of stable analogs of oxidized phospholipids with improved anti-inflammatory and barrier-protective properties.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-(410)-706-2578; Fax: +1-(410)-706-6952
| |
Collapse
|
13
|
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
|
14
|
The Protective Role of Heme Oxygenase-1 in Atherosclerotic Diseases. Int J Mol Sci 2019; 20:ijms20153628. [PMID: 31344980 PMCID: PMC6695885 DOI: 10.3390/ijms20153628] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. These products have anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-thrombotic properties. Although HO-1 is expressed at low levels in most tissues under basal conditions, it is highly inducible in response to various pathophysiological stresses/stimuli. HO-1 induction is thus thought to be an adaptive defense system that functions to protect cells and tissues against injury in many disease settings. In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. In animal models, a lack of HO-1 was shown to accelerate atherosclerosis, whereas HO-1 induction reduced atherosclerosis. It was also reported that HO-1 induction improved the cardiac function and postinfarction survival in animal models of heart failure or myocardial infarction. Recently, we and others examined blood HO-1 levels in patients with atherosclerotic diseases, e.g., coronary artery disease (CAD) and peripheral artery disease (PAD). Taken together, these findings to date support the notion that HO-1 plays a protective role against the progression of atherosclerotic diseases. This review summarizes the roles of HO-1 in atherosclerosis and focuses on the clinical studies that examined the relationships between HO-1 levels and atherosclerotic diseases.
Collapse
|
15
|
Fiorelli S, Porro B, Cosentino N, Di Minno A, Manega CM, Fabbiocchi F, Niccoli G, Fracassi F, Barbieri S, Marenzi G, Crea F, Cavalca V, Tremoli E, Eligini S. Activation of Nrf2/HO-1 Pathway and Human Atherosclerotic Plaque Vulnerability:an In Vitro and In Vivo Study. Cells 2019; 8:E356. [PMID: 30995787 PMCID: PMC6523494 DOI: 10.3390/cells8040356] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) induce nuclear factor erythroid 2-related factor 2 (Nrf2) activation as an adaptive defense mechanism, determining the synthesis of antioxidant molecules, including heme-oxygenase-1 (HO-1). HO-1 protects cells against oxidative injury, degrading free heme and inhibiting ROS production. HO-1 is highly expressed in macrophages during plaque growth. Macrophages are morpho-functionally heterogeneous, and the prevalence of a specific phenotype may influence the plaque fate. This heterogeneity has also been observed in monocyte-derived macrophages (MDMs), a model of macrophages infiltrating tissue. The study aims to assess oxidative stress status and Nrf2/HO-1 axis in MDM morphotypes obtained from healthy subjects and coronary artery disease (CAD) patients, in relation to coronary plaque features evaluated in vivo by optical coherence tomography (OCT). We found that MDMs of healthy subjects exhibited a lower oxidative stress status, lower Nrf2 and HO-1 levels as compared to CAD patients. High HO-1 levels in MDMs were associated with the presence of a higher macrophage content, a thinner fibrous cap, and a ruptured plaque with thrombus formation, detected by OCT analysis. These findings suggest the presence of a relationship between in vivo plaque characteristics and in vitro MDM profile, and may help to identify patients with rupture-prone coronary plaque.
Collapse
Affiliation(s)
| | - Benedetta Porro
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | | | | | | | | | - Giampaolo Niccoli
- Department of Cardiovascular & Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, I.R.C.C.S., Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Francesco Fracassi
- Department of Cardiovascular & Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, I.R.C.C.S., Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Simone Barbieri
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Giancarlo Marenzi
- Department of Cardiovascular & Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, I.R.C.C.S., Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Filippo Crea
- Department of Cardiovascular & Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, I.R.C.C.S., Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Viviana Cavalca
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Elena Tremoli
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Sonia Eligini
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| |
Collapse
|
16
|
Madrigal-Matute J, Martinez-Pinna R, Ramos-Mozo P, Blanco-Colio L, Moreno J, Tarin C, Burillo E, Fernandez-Garcia C, Egido J, Meilhac O, Michel JB, Martin-Ventura J. Erythrocytes, leukocytes and platelets as a source of oxidative stress in chronic vascular diseases: Detoxifying mechanisms and potential therapeutic options. Thromb Haemost 2017; 108:435-42. [DOI: 10.1160/th12-04-0248] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/21/2012] [Indexed: 12/15/2022]
Abstract
SummaryOxidative stress is involved in the chronic pathological vascular remodelling of both abdominal aortic aneurysm and occlusive atherosclerosis. Red blood cells (RBCs), leukocytes and platelets present in both, aneurysmal intraluminal thrombus and intraplaque haemorraghes, could be involved in the redox imbalance inside diseased arterial tissues. RBCs haemolysis may release the pro-oxidant haemoglobin (Hb), which transfers heme to tissue and low-density lipoproteins. Heme-iron potentiates molecular, cell and tissue toxicity mediated by leukocytes and other sources of reactive oxygen species (ROS). Polymorphonuclear neutrophils release myeloperoxidase and, along with activated platelets, produce superoxide mediated by NADPH oxidase, causing oxidative damage. In response to this pro-oxidant milieu, several anti-oxidant molecules of plasma or cell origin can prevent ROS production. Free Hb binds to haptoglobin (Hp) and once Hp-Hb complex is endocytosed by CD163, liberated heme is converted into less toxic compounds by heme oxygenase-1. Iron homeostasis is mainly regulated by transferrin, which transports ferric ions to other cells. Transferrin-bound iron is internalised via endocytosis mediated by transferrin receptor. Once inside the cell, iron is mainly stored by ferritin. Other non hemo-iron related antioxidant enzymes (e.g. superoxide dismutase, catalase, thioredoxin and peroxiredoxin) are also involved in redox modulation in vascular remodelling. Oxidative stress is a main determinant of chronic pathological remodelling of the arterial wall, partially linked to the presence of RBCs, leukocytes, platelets and oxidised fibrin within tissue and to the imbalance between pro-/anti-oxidant molecules. Understanding the complex mechanisms underlying redox imbalance could help to define novel potential targets to decrease atherothrombotic risk.
Collapse
|
17
|
Vogel ME, Idelman G, Konaniah ES, Zucker SD. Bilirubin Prevents Atherosclerotic Lesion Formation in Low-Density Lipoprotein Receptor-Deficient Mice by Inhibiting Endothelial VCAM-1 and ICAM-1 Signaling. J Am Heart Assoc 2017; 6:JAHA.116.004820. [PMID: 28365565 PMCID: PMC5532999 DOI: 10.1161/jaha.116.004820] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Numerous epidemiological studies support an inverse association between serum bilirubin levels and the incidence of cardiovascular disease; however, the mechanism(s) by which bilirubin may protect against atherosclerosis is undefined. The goals of the present investigations were to assess the ability of bilirubin to prevent atherosclerotic plaque formation in low‐density lipoprotein receptor‐deficient (Ldlr−/−) mice and elucidate the molecular processes underlying this effect. Methods and Results Bilirubin, at physiological concentrations (≤20 μmol/L), dose‐dependently inhibits THP‐1 monocyte migration across tumor necrosis factor α–activated human umbilical vein endothelial cell monolayers without altering leukocyte binding or cytokine production. A potent antioxidant, bilirubin effectively blocks the generation of cellular reactive oxygen species induced by the cross‐linking of endothelial vascular cell adhesion molecule 1 (VCAM‐1) or intercellular adhesion molecule 1 (ICAM‐1). These findings were validated by treating cells with blocking antibodies or with specific inhibitors of VCAM‐1 and ICAM‐1 signaling. When administered to Ldlr−/− mice on a Western diet, bilirubin (30 mg/kg intraperitoneally) prevents atherosclerotic plaque formation, but does not alter circulating cholesterol or chemokine levels. Aortic roots from bilirubin‐treated animals exhibit reduced lipid and collagen deposition, decreased infiltration of monocytes and lymphocytes, fewer smooth muscle cells, and diminished levels of chlorotyrosine and nitrotyrosine, without changes in VCAM‐1 or ICAM‐1 expression. Conclusions Bilirubin suppresses atherosclerotic plaque formation in Ldlr−/− mice by disrupting endothelial VCAM‐1‐ and ICAM‐1‐mediated leukocyte migration through the scavenging of reactive oxygen species signaling intermediaries. These findings suggest a potential mechanism for the apparent cardioprotective effects of bilirubin.
Collapse
Affiliation(s)
- Megan E Vogel
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Gila Idelman
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eddy S Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Disease Institute, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Stephen D Zucker
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
18
|
Hussain MS, Qureshi AI, Kirmani JF, Divani AA, Hopkins LN. Development of Vascular Biology over the past 10 Years: Heme Oxygenase-1 in Cardiovascular Homeostasis. J Endovasc Ther 2016; 11 Suppl 2:II32-42. [PMID: 15760262 DOI: 10.1177/15266028040110s616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The study of vascular biology has provided strong evidence for the role that free radical attack plays in the pathogenesis of cardiovascular diseases. The endothelial cell (EC) dysfunction that results from exposure to oxidative stresses, such as oxidized LDL, influences vascular cell gene expression, promoting smooth muscle cell (SMC) mitogenesis and apoptosis. These factors also play an important role in atherogenesis, which is attenuated by antioxidants. Thus, antioxidants are important to understanding the pathophysiology of cardiovascular diseases and to constructing an effective treatment strategy for these patients. Over the last decade, there has been a tremendous interest in the biology of heme oxygenase-1 (HO-1), which exhibits antioxidant effects in various forms of tissue injury. Moreover, the reaction is also the major source of carbon dioxide (CO) in the body, which is a physiologically important gaseous vasodilator that inhibits SMC proliferation. Thus, HO-1–derived products provide various mechanisms to maintain cardiovascular homeostasis. We review recent work on the cellular and molecular biological aspects of the HO/CO system in vascular pathophysiology.
Collapse
Affiliation(s)
- M Shazam Hussain
- Zeenat Qureshi Stroke Research Center, Department of Neurology and Neurosciences, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07101, USA
| | | | | | | | | |
Collapse
|
19
|
Abstract
Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.
Collapse
Affiliation(s)
- Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Abolfazl Zarjou
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Anupam Agarwal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
20
|
Lu Y, Hu J, Liu X. Bilirubin may limit heart ischemia and reperfusion injury in diabetic rats. Int J Cardiol 2016; 214:390. [DOI: 10.1016/j.ijcard.2016.03.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 03/27/2016] [Indexed: 10/22/2022]
|
21
|
Hormetic and anti-inflammatory properties of oxidized phospholipids. Mol Aspects Med 2016; 49:78-90. [DOI: 10.1016/j.mam.2016.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 12/15/2022]
|
22
|
Peterson SJ, Vanella L, Bialczak A, Schragenheim J, Li M, Bellner L, Shapiro JI, Abraham NG. Oxidized HDL and Isoprostane Exert a Potent Adipogenic Effect on Stem Cells: Where in the Lineage? ACTA ACUST UNITED AC 2016; 2. [PMID: 29430566 PMCID: PMC5807016 DOI: 10.16966/2472-6990.109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Stephen J Peterson
- Weill Cornell Medical College, Department of Medicine, New York Methodist Hospital, Brooklyn, NY 11215, USA
| | - Luca Vanella
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA.,Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Angelica Bialczak
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Joseph Schragenheim
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Ming Li
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Lars Bellner
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Joseph I Shapiro
- Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| | - Nader G Abraham
- Departments of Medicine & Pharmacology, New York Medical College, Valhalla, NY 10595, USA.,Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA
| |
Collapse
|
23
|
Ikwuobe J, Bellary S, Griffiths HR. Innovative biomarkers for predicting type 2 diabetes mellitus: relevance to dietary management of frailty in older adults. Biogerontology 2016; 17:511-27. [PMID: 26897532 DOI: 10.1007/s10522-016-9634-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 01/18/2016] [Indexed: 01/01/2023]
Abstract
Type 2 diabetes mellitus (T2DM) increases in prevalence in the elderly. There is evidence for significant muscle loss and accelerated cognitive impairment in older adults with T2DM; these comorbidities are critical features of frailty. In the early stages of T2DM, insulin sensitivity can be improved by a "healthy" diet. Management of insulin resistance by diet in people over 65 years of age should be carefully re-evaluated because of the risk for falling due to hypoglycaemia. To date, an optimal dietary programme for older adults with insulin resistance and T2DM has not been described. The use of biomarkers to identify those at risk for T2DM will enable clinicians to offer early dietary advice that will delay onset of disease and of frailty. Here we have used an in silico literature search for putative novel biomarkers of T2DM risk and frailty. We suggest that plasma bilirubin, plasma, urinary DPP4-positive microparticles and plasma pigment epithelium-derived factor merit further investigation as predictive biomarkers for T2DM and frailty risk in older adults. Bilirubin is screened routinely in clinical practice. Measurement of specific microparticle frequency in urine is less invasive than a blood sample so is a good choice for biomonitoring. Future studies should investigate whether early dietary changes, such as increased intake of whey protein and micronutrients that improve muscle function and insulin sensitivity, affect biomarkers and can reduce the longer term complication of frailty in people at risk for T2DM.
Collapse
Affiliation(s)
- John Ikwuobe
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Birmingham, B4 7ET, UK
| | - Srikanth Bellary
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Birmingham, B4 7ET, UK
| | - Helen R Griffiths
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
24
|
Huang SS, Chan WL, Leu HB, Huang PH, Lin SJ, Chen JW. Serum bilirubin levels predict future development of metabolic syndrome in healthy middle-aged nonsmoking men. Am J Med 2015; 128:1138.e35-41. [PMID: 25912203 DOI: 10.1016/j.amjmed.2015.04.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 04/05/2015] [Accepted: 04/11/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Despite epidemiologic research demonstrating an inverse relationship between serum bilirubin levels and the prevalence of metabolic syndrome, prospective data on serum bilirubin as a predictor of incident metabolic syndrome are limited. METHODS Serum bilirubin was examined as a risk marker for incident metabolic syndrome in a prospective study of 468 Taiwanese middle-aged men who were free of metabolic syndrome and other systemic diseases at baseline. These subjects were followed up in annual health examinations between 2001 and 2009 for the development of metabolic syndrome, which was defined according to unified criteria set by several major organizations. RESULTS Among the study subjects, 377 were nonsmokers and 91 were current smokers. All individuals were then stratified into 3 groups according to their baseline serum bilirubin levels (low, normal, and high). During a mean follow-up period of 7.58 years, 66 subjects developed metabolic syndrome. The incidence of metabolic syndrome was significantly reduced in the high-bilirubin group compared with the low-bilirubin group (6.4% vs 22.4%, P < .001). Multivariate Cox regression analysis revealed that the hazard ratio for incident metabolic syndrome between the highest and lowest tertiles of serum bilirubin levels was 0.246 (95% confidence interval 0.120-0.503). However, although it remained clearly evident in nonsmokers, the inverse correlation was attenuated in current smokers. CONCLUSIONS Increased serum bilirubin was associated with a reduced future risk of metabolic syndrome in apparently healthy middle-aged, nonsmoking men. Our findings support the predictive role of serum total bilirubin for future development of metabolic syndrome.
Collapse
Affiliation(s)
- Shao-Sung Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Healthcare and Service Center, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Leong Chan
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Healthcare and Service Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Bang Leu
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Healthcare and Service Center, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jaw-Wen Chen
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
25
|
Zhang ZY, Guan J, Li H, Zhou ZQ, Zhou GW. Heme Oxygenase-1 Promoter Polymorphism Protects Liver Allograft. Indian J Surg 2015; 78:14-19. [PMID: 27186034 DOI: 10.1007/s12262-015-1309-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/19/2015] [Indexed: 11/24/2022] Open
Abstract
Heme oxygenase-1 has been identified to protect allograft from ischemia/reperfusion and immunologic rejection. Activity of heme oxygenase-1 is regulated by a guanine-thymine dinucleotide length polymorphism in the heme oxygenase-1 gene promoter. In this study, we aimed to explore the impact of the heme oxygenase-1 gene promoter polymorphism of donors and recipients on the orthotopic liver graft function after transplantation. Sixty recipients and their accompanying donors of orthotopic liver allografts were included retrospectively in this study. Heme oxygenase-1 gene promoter polymorphism was assessed using genomic DNA isolated from cryopreserved splenocytes or peripheral blood mononuclear cells and analyzed by genetic analyzer. Small allele of the donor heme oxygenase-1 gene polymorphism significantly prolonged the graft survival (p = 0.017). Recipients of allografts from a class of small-allele carrier had significantly lower serum total bilirubin compared with recipients of a nonclass small-allele donor liver (p < 0.01). Additionally, in recipients of small-carrier allografts, cold ischemia time (<10 h or ≥10 h) did not affect the total bilirubin significantly. Our study suggested a protective function of donor-derived heme oxygenase-1 gene promoter polymorphism on orthotopic liver allograft function after transplantation.
Collapse
Affiliation(s)
- Zheng-Yun Zhang
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, 200233 Shanghai, China
| | - Jiao Guan
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, 200233 Shanghai, China
| | - Hao Li
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, 200233 Shanghai, China
| | - Zun-Qiang Zhou
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, 200233 Shanghai, China
| | - Guang-Wen Zhou
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, 200233 Shanghai, China
| |
Collapse
|
26
|
Kobayashi A, Mizukami H, Sakamoto N, Yamaki T, Kunii H, Nakazato K, Takeishi Y. ENDOGENOUS CARBON MONOXIDE CONCENTRATION IN BLOOD ELEVATES IN ACUTE CORONARY SYNDROME OF NONSMOKER POPULATION. Fukushima J Med Sci 2015; 61:72-8. [PMID: 26135664 DOI: 10.5387/fms.2015-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Carbon monoxide (CO) was previously only considered as a highly toxic pollutant since it binds to hemoglobin with high affinity. Recently, however, it has been recognized as a signaling molecule with regulatory roles in many physiological and pathophysiological processes within the cardiovascular system. The aim of this study was to clarify the behavior of CO in patients with acute coronary syndrome (ACS). METHODS We assessed 235 patients with suspected ACS, 98 smokers (88 male, 62 ± 14 years) and 137 nonsmokers (77 male, 72 ± 13 years), who had undergone emergent cardiac catheterization and blood sampling for calculation of carboxyhemoglobin (COHb). Patients were categorized into 4 groups: smoking patients with ACS (n=77), smoking patients without ACS (n=21), non-smoking patients with ACS (n=97), and non-smoker patients without ACS (n=40). We investigated whether biomarkers were related to COHb levels. RESULTS LogCOHb was significantly higher in the smoking patients compared to non-smoking patients (0.30 ± 0.12 vs. 0.45 ± 0.18, P < 0.01). Interestingly, among the non-smoking patients, COHb was increased in the ACS patients compared to the non ACS patients (0.31 ± 0.12 vs. 0.25 ± 0.12 P < 0.01). In contrast, among the smoking patients, there was no difference in COHb between the ACS and non-ACS patients (0.45 ± 0.18 vs. 0.44 ± 0.18, n.s.). There were no correlations between COHb and any of the biomarkers. CONCLUSIONS These results suggest that endogenous CO may be useful to assess the risk of cardiovascular stress.
Collapse
Affiliation(s)
- Atsushi Kobayashi
- Department of Cardiology and Hematology, Fukushima Medical University
| | | | | | | | | | | | | |
Collapse
|
27
|
Daenen KEL, Martens P, Bammens B. Association of HO-1 (GT)n Promoter Polymorphism and Cardiovascular Disease: A Reanalysis of the Literature. Can J Cardiol 2015; 32:160-8. [PMID: 26483091 DOI: 10.1016/j.cjca.2015.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/27/2015] [Accepted: 06/11/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Heme-oxygenase 1 (HO-1), an inducible heme-degrading enzyme, has antiatherogenic effects through its enzymatic end products. HO-1 gene expression is modulated by a guanidine thymidine dinucleotide ([GT]n) repeat polymorphism in the promoter region. Shorter repeats with (GT)n < 25 are associated with higher inducibility and activity of HO-1. METHODS We performed a systematic review of all literature from 1997 to 2013 on the association of the HO-1 (GT)n and cardiovascular disease (CVD). On the basis of predefined criteria (patient characteristics, genotype data format, allelic distribution, repeat length cutoff) 41 articles were selected. Patients were redistributed into 4 homogeneous subpopulations: patients with CVD (CVD group), patients without CVD (nonCVD), 'controls' with unknown cardiovascular status (unspecified) and children younger than 20 years of age (unselected). Genotype distributions (homozygous short [SS] or long [LL], and heterozygous) of the 4 patient categories were compared and odds ratios (ORs) for CVD were calculated using logistic regression analysis. RESULTS Overall, the proportion of the SS genotype was lower in CVD compared with nonCVD and unspecified. The ORs for CVD was highest in patients carrying the LL genotype (OR LL vs SS, 1.769 [95% confidence interval, 1.594-1.963]). Furthermore, genotype distribution differed between Caucasian and Asian individuals, the latter having a much higher proportion of the SS genotype (22% vs 11%). CONCLUSIONS This review of the available literature on the epidemiological association between the HO-1 (GT)n repeat polymorphism and CVD supports the presumed protective effects of HO-1. The second but probably even more relevant finding of our review is that racial disparities in HO-1 (GT)n repeat length distribution exist and might influence the associations of the genotype with CVD status.
Collapse
Affiliation(s)
- Kristien E L Daenen
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Martens
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Bert Bammens
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
28
|
Li Y, Guo Y, Chen Y, Wang Y, You Y, Yang Q, Weng X, Li Q, Zhu X, Zhou B, Liu X, Gong Z, Zhang R. Establishment of an interleukin-1β-induced inflammation-activated endothelial cell-smooth muscle cell-mononuclear cell co-culture model and evaluation of the anti-inflammatory effects of tanshinone IIA on atherosclerosis. Mol Med Rep 2015; 12:1665-76. [PMID: 25936371 PMCID: PMC4464412 DOI: 10.3892/mmr.2015.3668] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 02/19/2015] [Indexed: 02/02/2023] Open
Abstract
Increasing evidence supports the hypothesis that inflammatory reactions serves an important function in the formation, progression and plaque rupture of atherosclerosis. Interleukin (IL)-1 primarily induces inflammation and is closely associated with the inflammatory environment and the formation of atherosclerosis. The present study aimed to establish an in vitro model for the evaluation of drug efficacy in the intervention of atherosclerosis from the inflammatory perspective, and to observe the anti-inflammatory effects of tanshinone IIA and andrographolide on atherosclerosis. The IL-1β-induced inflammation-activated endothelial cell (EC)-smooth muscle cell (SMC)-mononuclear cell (MC) co-culture model was established, based on the changes in a series of atherosclerosis-associated inflammatory markers secreted by ECs and SMCs. The expression of connexin in ECs, adhesion of MCs and changes in inflammatory signalling molecules were selected as evaluation indices for the inflammatory microenvironment of atherosclerosis. The use of this model revealed that tanshinone IIA exhibited significant efficacy against atherosclerosis and its inflammatory reactions. Inflammatory reactions were regarded as the primary mechanism underlying atherosclerosis. The established model simulated a series of relevant changes in the arterial wall under the inflammatory cytokines with oxidized low-density lipoprotein during the atherosclerotic process. The present study presented a reliable method for the identification of drugs with potential anti-inflammatory activity in atherosclerosis, for investigating the mechanisms of action, considering the improvement of the inflammatory state and the increase in plaque stability observed.
Collapse
Affiliation(s)
- Yujie Li
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yan Guo
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Ying Chen
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yajie Wang
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yun You
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Qing Yang
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Xiaogang Weng
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Qi Li
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Xiaoxin Zhu
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Bingbing Zhou
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Xucen Liu
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Zaipeng Gong
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Ruijie Zhang
- Department of Pharmacokinetics of Chinese Materia Medica, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| |
Collapse
|
29
|
Levitt DG, Levitt MD. Carbon monoxide: a critical quantitative analysis and review of the extent and limitations of its second messenger function. Clin Pharmacol 2015; 7:37-56. [PMID: 25750547 PMCID: PMC4348054 DOI: 10.2147/cpaa.s79626] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Endogenously produced carbon monoxide (CO) is commonly believed to be a ubiquitous second messenger involved in a wide range of physiological and pathological responses. The major evidence supporting this concept is that CO is produced endogenously via heme oxygenase-catalyzed breakdown of heme and that experimental exposure to CO alters tissue function. However, it remains to be conclusively demonstrated that there are specific receptors for CO and that endogenous CO production is sufficient to alter tissue function. Unlike other signaling molecules, CO is not significantly metabolized, and it is removed from cells solely via rapid diffusion into blood, which serves as a near infinite sink. This non-metabolizable nature of CO renders the physiology of this gas uniquely susceptible to quantitative modeling. This review analyzes each of the steps involved in CO signaling: 1) the background CO partial pressure (PCO) and the blood and tissue CO binding; 2) the affinity of the putative CO receptors; 3) the rate of endogenous tissue CO production; and 4) the tissue PCO that results from the balance between this endogenous CO production and diffusion to the blood sink. Because existing data demonstrate that virtually all endogenous CO production results from the routine "housekeeping" turnover of heme, only a small fraction can play a signaling role. The novel aspect of the present report is to demonstrate via physiological modeling that this small fraction of CO production is seemingly insufficient to raise intracellular PCO to the levels required for the conventional, specific messenger receptor activation. It is concluded that the many physiological alterations observed with exogenous CO administration are probably produced by the non-specific CO inhibition of cytochrome C oxidase activity, with release of reactive oxygen species (ROS) and that this ROS signaling pathway is a potential effector mechanism for endogenously produced CO.
Collapse
Affiliation(s)
- David G Levitt
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Michael D Levitt
- Research Service, Veterans Affairs Medical Center, Minneapolis, MN, USA
| |
Collapse
|
30
|
Zuniga MC, White SLP, Zhou W. Design and utilization of macrophage and vascular smooth muscle cell co-culture systems in atherosclerotic cardiovascular disease investigation. Vasc Med 2014; 19:394-406. [DOI: 10.1177/1358863x14550542] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherosclerotic cardiovascular disease has been acknowledged as a chronic inflammatory condition. Monocytes and macrophages lead the inflammatory pathology of atherosclerosis whereas changes in atheromatous plaque thickness and matrix composition are attributed to vascular smooth muscle cells. Because these cell types are key players in atherosclerosis progression, it is crucial to utilize a reliable system to investigate their interaction. In vitro co-culture systems are useful platforms to study specific molecular mechanisms between cells. This review aims to summarize the various co-culture models that have been developed to investigate vascular smooth muscle cell and monocyte/macrophage interactions, focusing on the monocyte/macrophage effects on vascular smooth muscle cell function.
Collapse
Affiliation(s)
- Mary C Zuniga
- Surgical Services, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sharla L Powell White
- Division of Vascular Surgery, School of Medicine, Stanford University, Stanford, CA, USA
- Cardiovascular Institute, School of Medicine, Stanford University, Stanford, CA, USA
| | - Wei Zhou
- Surgical Services, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Vascular Surgery, School of Medicine, Stanford University, Stanford, CA, USA
- Cardiovascular Institute, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
31
|
Atorvastatin represses the angiotensin 2-induced oxidative stress and inflammatory response in dendritic cells via the PI3K/Akt/Nrf 2 pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:148798. [PMID: 25110549 PMCID: PMC4106155 DOI: 10.1155/2014/148798] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs), which are highly proficient antigen-presenting cells, play a complex role in both the initiation and progression of atherosclerosis. We tested the hypothesis that the anti-inflammatory and antioxidant effects of atorvastatin may be partly mediated by the phosphatidylinositol 3-kinase/protein kinase B/transcription factor nuclear factor-erythroid 2-related factor 2 (PI3K/Akt/Nrf 2) pathway via the attenuation of DC maturation, thus reducing the inflammatory and oxidative stress responses. This study showed that angiotensin 2 (Ang 2) induced the maturation of DCs, stimulated CD83, CD40, CD80, and CD86 expression, and increased the secretion of IL-12p70, IL-6, and TNF-α. These effects were suppressed by atorvastatin. Atorvastatin also lowered the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), counteracting their initial increases in response to Ang 2 stimulation. Atorvastatin activated Nrf 2 via the PI3K/Akt pathway and thereby promoted Nrf 2 translocation from the cytoplasm to the nucleus in bone marrow-derived dendritic cells (BMDCs), a process that was reversed by the PI3K inhibitor LY294002. Therefore, the regulation of Nrf 2 expression by the PI3K/Akt pathway plays an important role in the regulation of the statin-mediated antioxidant and anti-inflammatory responses in DCs.
Collapse
|
32
|
Abstract
This review highlights the protective roles of bilirubin against the atherosclerotic process. Bilirubin belongs to the superfamily of tetrapyrrolic compounds formed during heme catabolism. Although for decades bilirubin was considered to be a harmful waste product, recent epidemiologic studies have shown that serum bilirubin levels have consistently been inversely associated with cardiovascular disease (CVD), as well as cardiovascular risk factors such as metabolic syndrome and diabetes. These clinical studies are supported by in vitro and in vivo experimental data and have demonstrated that bilirubin not only has an ability to scavenge overproduced reactive oxygen species and inhibit vascular smooth muscle cell proliferation but, additionally, has anti-inflammatory effects. In this review, we will discuss the inverse association of serum bilirubin and CVD and cardiovascular risk factors established in various clinical studies. We also review detailed experimental studies about the effect of bilirubin on atherosclerotic processes. In vitro, animal and human studies have proved that bilirubin inhibits oxidation of cholesterol which is an important step of atherosclerosis. Bilirubin attenuates chemotactic activity of monocytes and strongly inhibits adhesion of leukocytes to venule and production of pro-inflammatory cytokines. Bilirubin has inhibited serum-driven smooth muscle cell cycle progression at the G1 phase. Lastly, we will discuss briefly the influence of bilirubin on lipoprotein composition and endothelial dysfunction.
Collapse
Affiliation(s)
- Seung Joo Kang
- Department of Internal Medicine, Healthcare Research Institute, Seoul National University, Hospital Healthcare System Gangnam Center , Seoul , Korea
| | | | | |
Collapse
|
33
|
Zheng H, Li Y, Xie N. Association of serum total bilirubin levels with diastolic dysfunction in heart failure with preserved ejection fraction. Biol Res 2014; 47:7. [PMID: 25027460 PMCID: PMC4101732 DOI: 10.1186/0717-6287-47-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/18/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Left ventricular diastolic dysfunction is one of the main characteristics of heart failure patients with a preserved left ventricular ejection fraction. As bilirubin is regarded as an important endogenous antioxidant molecule, serum total bilirubin levels were compared between heart failure patients with a preserved left ventricular ejection fraction and normal controls in this study. We recruited 327 heart failure patients with a preserved left ventricular ejection fraction and 200 healthy controls. Patients were divided into 4 subgroups by their comprehensive echocardiographic manifestations, 1-mild, 2-moderate, 3-severe (reversible restrictive), 4-severe (fixed restrictive). Total bilirubin levels were compared using stepwise multiple regressions adjusted for selected factors. RESULTS After adjusting for gender, age, smoking, systolic blood pressure, diastolic blood pressure, total cholesterol and triglyceride, serum total bilirubin levels were significantly lower in the heart failure group compared with the control group (P < 0.01). Patients in the subgroup (4-severe) showed significantly (P < 0.05) lower levels of total bilirubin when compared with the subgroup (1-mild). CONCLUSIONS TB level was negatively correlated with left ventricular diastolic dysfunction in heart failure patients with a preserved left ventricular ejection fraction, which might provide a new insight into the complicated mechanisms of heart failure with a preserved left ventricular ejection fraction.
Collapse
|
34
|
Li B, Lee YJ, Kim YC, Yoon JJ, Lee SM, Lee YP, Kang DG, Lee HS. Sauchinone from Saururus chinensis protects vascular inflammation by heme oxygenase-1 induction in human umbilical vein endothelial cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:101-108. [PMID: 24035224 DOI: 10.1016/j.phymed.2013.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/09/2013] [Accepted: 08/09/2013] [Indexed: 06/02/2023]
Abstract
Sauchinone, a diastereomeric lignan isolated from the roots of Saururus chinensis (LOUR.) BAILL. (Saururaceae), is reported to exert a variety of biological activities such as hepatoprotective, anti-inflammatory actions and inhibitory effects on bone resorption. In this study, we investigated the effect of sauchinone in suppressing cell adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1) and intracellular adhesion molecule-1 (ICAM-1) expression in high glucose stimulated human umbilical vein endothelial cells (HUVEC). Sauchinone inhibited the phosphorylation and degradation of IκB-α, as well as the nuclear translocation of nuclear factor kappa B (NF-κB) p65 caused by the stimulation of high glucose. In addition, sauchinone induced heme oxygenase (HO)-1 expression through nuclear translocation of nuclear factor E2-related factor 2 in HUVEC. The effects of sauchinone on the high glucose-induced expression of VCAM-1 and ICAM-1 and nuclear translocation of NF-κB p65 were partially reversed by transfection of the cells with HO-1 siRNA. These findings suggest that sauchinone-induced HO-1 expression plays a key role in the vascular protective effects of sauchinone in HUVEC.
Collapse
Affiliation(s)
- Bin Li
- Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea
| | - Yun Jung Lee
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea
| | - Youn Chul Kim
- Standardized Material Bank for New Botanical Drugs; College of Pharmacy, Wonkwang University, Iksan 570-749, Republic of Korea
| | - Jung Joo Yoon
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea
| | - So Min Lee
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea
| | - Yong Pyo Lee
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea
| | - Dae Gill Kang
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea.
| | - Ho Sub Lee
- College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Shinyong-dong, Iksan, Jeonbuk 570-749, Republic of Korea; Hanbang Body-fluid Research Center, Wonkwang University, Shinyong-dong, Iksan Jeonbuk, 570-749, Republic of Korea.
| |
Collapse
|
35
|
Yunoki K, Inoue T, Sugioka K, Nakagawa M, Inaba M, Wada S, Ohsawa M, Komatsu R, Itoh A, Haze K, Yoshiyama M, Becker AE, Ueda M, Naruko T. Association between hemoglobin scavenger receptor and heme oxygenase-1-related anti-inflammatory mediators in human coronary stable and unstable plaques. Hum Pathol 2013; 44:2256-65. [PMID: 23850497 DOI: 10.1016/j.humpath.2013.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 04/05/2013] [Accepted: 05/08/2013] [Indexed: 10/26/2022]
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. In vitro studies have shown that binding of hemoglobin to hemoglobin scavenger receptor (CD163) induces HO-1 and the anti-inflammatory mediator interleukin (IL)-10. We immunohistochemically examined the relationship between CD163 expression in macrophages and intraplaque hemorrhage, HO-1, IL-10, and ferritin using coronary atherectomy specimens from patients with stable (SAP) or unstable angina pectoris (UAP). A total of 67 patients underwent atherectomy for SAP (n = 33) or UAP (n = 34). Samples were stained with antibodies against smooth muscle cells, macrophages, glycophorin-A (a protein specific to erythrocyte membranes), CD163, HO-1, IL-10, and ferritin. To identify cell types of HO-1-positive cells, double immunostaining was also performed. Double immunostaining for HO-1 and macrophages revealed that the vast majority of HO-1-positive cells were macrophages. Morphometric analysis demonstrated that CD163-positive macrophage score and the percentage of glycophorin-A-, HO-1-, IL-10-, and ferritin-positive areas were significantly higher in UAP than in SAP patients (CD163, P < .005; glycophorin-A, P < .0001; HO-1, P < .0001; IL-10, P < .005; ferritin, P = .0001). Moreover, CD163-positive macrophage score was positively associated with the percentage of glycophorin-A-, HO-1-, IL-10-, and ferritin-positive areas (glycophorin-A, r = 0.60, P < .0001; HO-1, r = 0.67, P < .0001; IL-10, r = 0.45, P < .0005; ferritin, r = 0.61, P < .0001). These findings suggest that enhanced expression of HO-1 and HO-1-related atheroprotective molecules plays an important role in exerting anti-inflammatory, antioxidant, and scavenging functions, which could contribute to plaque stabilization.
Collapse
Affiliation(s)
- Kei Yunoki
- Department of Cardiology, Osaka City General Hospital, Osaka 534-0021, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kao TW, Chou CH, Wang CC, Chou CC, Hu J, Chen WL. Associations between serum total bilirubin levels and functional dependence in the elderly. Intern Med J 2013; 42:1199-207. [PMID: 22032210 DOI: 10.1111/j.1445-5994.2011.02620.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 10/20/2011] [Indexed: 11/26/2022]
Abstract
BACKGROUND Many studies support the role of bilirubin as a cytoprotector in chronic inflammatory diseases, such as stroke and atherosclerosis. AIM To investigate the relationship between serum total bilirubin levels and functional dependence in older adults. METHODS Data from the National Health and Nutrition Examination Survey (1999-2002) pertaining to 2235 old adults were analysed. All participants had given a household interview, providing information of five major domains on self-reported functional status (activities of daily living, instrumental activities of daily living, leisure and social activities, lower extremity mobility and general physical activities), had completed serum total bilirubin measurement, and a questionnaire regarding personal health. Poor performance was defined as experiencing difficulty with one or more items in a given domain. Functional dependence was defined as having three or more poor performances in the five major domains. Multiple logistic regression was performed together with quartile-based stratified odds ratio (OR) comparison and trend tests. RESULTS The OR of functional dependence for each standard deviation increment in the serum total bilirubin level was 0.56 (P = 0.002). After additional adjustment, the inverse association remained essentially unchanged. In quartile-based analysis, participants with higher quartiles of serum total bilirubin tended to have lower ORs of functional dependence. The trends of lower likelihood of functional dependence across increasing quartiles of the serum total bilirubin level were statistically significant (P < 0.05 for all trends). CONCLUSIONS Higher serum total bilirubin levels were associated with lower likelihood of functional dependence in older adults.
Collapse
Affiliation(s)
- T W Kao
- Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan; School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Liu Q, Wang B, Yin Y, Chen G, Wang W, Gao X, Wang P, Zhou H. Overexpressions of HO-1/HO-1G143H in C57/B6J mice affect melanoma B16F10 lung metastases rather than change the survival rate of mice-bearing tumours. Exp Biol Med (Maywood) 2013; 238:696-704. [PMID: 23918881 DOI: 10.1177/1535370213490628] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is often upregulated in tumour tissues and endows tumour cells with cytoprotection and antiapoptosis. It is worthy of note that some people show higher activity of HO-1 and some anti-cancer therapies could induce HO-1 expression in normal tissues, but the effect of HO-1 of normal tissues on tumours among these people remains unknown. To assess the effect of HO-1 of normal tissues on tumour progressiveness, we investigated the growth, metastasis and angiogenic potential of murine melanoma B16F10 cells in transgenic mice overexpressing HO-1 and its negative dominant mutant HO-1G143H, respectively. The results demonstrated that neither overexpression of HO-1 nor overexpression of HO-1G143H in normal tissues could significantly change the survival rate of tumour-bearing mice, but HO-1 overexpression could inhibit lung metastases and HO-1G143H could significantly promote lung metastases. Meanwhile, the leukocytes infiltration was reduced and angiogenesis was promoted in tumours in mice overexpressing HO-1, but the opposite was true in mice overexpressing HO-1G143H. Our findings suggested that overexpression of HO-1 might be conducive to patients bearing melanoma metastasis.
Collapse
Affiliation(s)
- Qingjun Liu
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Bo Wang
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Yujing Yin
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Gan Chen
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Peng Wang
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing 100071, China
| | - Hong Zhou
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
38
|
Intermittent Hypoxia-Induced NF-κB and HO-1 Regulation in Human Endothelial EA.hy926 Cells. Cell Biochem Biophys 2012; 66:431-41. [DOI: 10.1007/s12013-012-9491-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Noh JS, Park CH, Tanaka T, Yokozawa T. 7-O-galloyl-D-sedoheptulose attenuates oxidative stress-induced diabetic injury via decreasing expression of nuclear factor-κB- and apoptosis-related protein in the liver. Biol Pharm Bull 2012; 35:950-6. [PMID: 22687537 DOI: 10.1248/bpb.35.950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study was conducted to examine whether 7-O-galloyl-D-sedoheptulose (GS) has an ameliorative effect on diabetic alterations such as oxidative stress, inflammation, and apoptosis in the liver of type 2 diabetic db/db mice. GS was administered at 20 or 100 mg/kg body weight per day for 6 weeks to db/db mice, and its effect was compared with vehicle-treated db/db and m/m mice. In the serum and hepatic tissue, biochemical factors and protein expressions associated with nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, inflammation, and apoptosis were examined. As a result, GS administration to type 2 diabetic mice lowered serum and hepatic oxidative stress through the reduction of reactive oxygen species and lipid peroxidation. These results were derived, at least in part, from attenuating the expression of NADPH oxidase subunit proteins, Nox-4 and p22(phox). In the diabetic condition, augmented nuclear factor (NF)-E2-related factor 2 and heme oxygenase-1 were reduced with a decrease in oxidative stress on GS treatment. Furthermore, in the GS-treated group, NF-kappa B-related pro-inflammatory factors and pro-apoptotic protein expressions were alleviated in the hepatic tissue. Taking these into consideration, our findings support the therapeutic evidence for GS ameliorating the development of diabetic complications via regulating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Jeong Sook Noh
- Institute of Natural Medicine, University of Toyama, Japan
| | | | | | | |
Collapse
|
40
|
Lönn ME, Dennis JM, Stocker R. Actions of "antioxidants" in the protection against atherosclerosis. Free Radic Biol Med 2012; 53:863-84. [PMID: 22664312 DOI: 10.1016/j.freeradbiomed.2012.05.027] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 05/05/2012] [Accepted: 05/16/2012] [Indexed: 02/07/2023]
Abstract
This review addresses the role of oxidative processes in atherosclerosis and its resulting cardiovascular disease by focusing on the outcome of antioxidant interventions. Although there is unambiguous evidence for the presence of heightened oxidative stress and resulting damage in atherosclerosis, it remains to be established whether this represents a cause or a consequence of the disease. This critical question is complicated further by the increasing realization that oxidative processes, including those related to signaling, are part of normal cell function. Overall, the results from animal interventions suggest that antioxidants provide benefit neither generally nor consistently. Where benefit is observed, it appears to be achieved at least in part via modulation of biological processes such as increase in nitric oxide bioavailability and induction of protective enzymes such as heme oxygenase-1, rather than via inhibition of oxidative processes and lipid oxidation in the arterial wall. Exceptions to this may be situations of multiple/excessive stress, the relevance of which for humans is not clear. This interpretation is consistent with the overall disappointing outcome of antioxidant interventions in humans and can be rationalized by the spatial compartmentalization of cellular oxidative signaling and/or damage, complex roles of oxidant-producing enzymes, and the multifactorial nature of atherosclerosis.
Collapse
Affiliation(s)
- Maria E Lönn
- Centre for Vascular Research, School of Medical Sciences (Pathology), and Bosch Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | | | | |
Collapse
|
41
|
Huang SS, Huang PH, Wu TC, Chen JW, Lin SJ. Association of serum bilirubin with contrast-induced nephropathy and future cardiovascular events in patients undergoing coronary intervention. PLoS One 2012; 7:e42594. [PMID: 22880046 PMCID: PMC3412818 DOI: 10.1371/journal.pone.0042594] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/09/2012] [Indexed: 11/18/2022] Open
Abstract
Objectives Enhanced reactive oxygen species formation within the kidney following the administration of contrast media may play a key role in the development of contrast-induced nephropathy (CIN). Bilirubin has emerged as an important endogenous antioxidant molecule. This study was undertaken to determine whether bilirubin is associated with CIN and future cardiovascular events in patients undergoing coronary intervention. Methods Totally, 544 consecutive patients received coronary intervention were enrolled. All patients were followed up for at least 3 years or until the occurrence of a major event. The primary endpoint was CIN, defined as a rise in serum creatinine (SCr) of 0.5 mg/dl or a 25% increase from the baseline value within 48 hours after the procedure. The secondary endpoint was the combined occurrence of major adverse cardiovascular events (MACE), including death, nonfatal myocardial infarction, and ischemic stroke. Results Overall, CIN occurred in 85 (15.6%) patients. All patients were stratified into 3 groups (low/normal/high) according to the serum bilirubin levels. In a multivariate logistic analysis, the odds ratio for CIN with low-bilirubin levels relative to high-bilirubin levels was 11.82 (95% CI, 3.25–43.03). By Cox regression analysis, serum bilirubin levels was an independent predictor of MACE in patients undergoing coronary intervention (low vs. high hazard ratio 2.26; 95% CI, 1.05–4.90). Conclusions CIN is a serious complication of coronary intervention. Higher serum bilirubin concentrations were associated with lower risk of CIN and fewer cardiovascular events. The development of interventions that promote bilirubin levels may be a potential target to reduce CIN and future MACE in patients undergoing coronary intervention.
Collapse
Affiliation(s)
- Shao-Sung Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| | - Tao-Cheng Wu
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
42
|
Chen M, Zhou L, Ding H, Huang S, He M, Zhang X, Cheng L, Wang D, Hu FB, Wu T. Short (GT) ( n ) repeats in heme oxygenase-1 gene promoter are associated with lower risk of coronary heart disease in subjects with high levels of oxidative stress. Cell Stress Chaperones 2012; 17:329-38. [PMID: 22120665 PMCID: PMC3312960 DOI: 10.1007/s12192-011-0309-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 10/20/2011] [Accepted: 10/21/2011] [Indexed: 01/30/2023] Open
Abstract
Although (GT) ( n ) repeats in heme oxygenase-1 (HO-1) promoter may modulate gene transcriptional activity, the association between (GT) ( n ) repeats polymorphism and risk of coronary heart disease (CHD) from different levels of oxidative stress (OS) is unknown. We determined the allelic frequencies of (GT) ( n ) repeats in the HO-1 gene promoter and plasma malonaldehyde (MDA) as biomarkers of OS in 2,298 pairs of CHD patients and controls in the Chinese population. Furthermore, we measured MDA in culture mediums and HO-1 expressions levels in cell lysates of endothelial cells carrying various (GT) ( n ) genotypes under different concentrations of H(2)O(2). Compared with L/L genotype (>25 repeats) carriers, the adjusted odd ratios for S/S genotype (≤25 repeats) in subjects with different levels of OS (MDA < 1.83, 1.83-2.91, >2.91 μmol/L) were 1.06 (95%CI, 0.75 to 1.49), 0.79 (95%CI, 0.55 to 1.12), and 0.60 (95%CI, 0.44 to 0.81), respectively (P (interaction) = 0.002). In biological experiments, compared with endothelial cells carrying L/L genotype, cells with S/S genotype did not have a significantly higher HO-1 expression under 0 μmol/L H(2)O(2), but displayed a significantly higher HO-1 expression under 50 μmol/L H(2)O(2) (P (interaction) = 0.003). S/S genotype in HO-1 gene promoter is associated with a lower risk of CHD in subjects with higher levels of OS, because under conditions of high OS, the S/S genotype has higher levels of HO-1, an antioxidant.
Collapse
Affiliation(s)
- Mu Chen
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Li Zhou
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Hu Ding
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030 China
| | - Suli Huang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Meian He
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Xiaomin Zhang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Longxian Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 32 Xinhua Rd, Wuhan, Hubei 430030 China
| | - Daowen Wang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030 China
| | - Frank B. Hu
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, Boston, MA 02115 USA
| | - Tangchun Wu
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| |
Collapse
|
43
|
Abstract
Oxidant stress plays an important role in the pathogenesis of atherosclerosis. In the late 1980s, biological studies demonstrated that oxygen-free radicals oxidize low-density lipoprotein-cholesterol, resulting in the creation of foam cells and inciting the cascade of biological events that ultimately result in the formation of atherosclerosis. In vitro studies showed the ability of antioxidant vitamins to scavenge free radicals and block the oxidation of low-density lipoprotein. This data was supported in vivo by early observational studies suggesting the benefit of antioxidants, particularly vitamin E, in the prevention of coronary artery disease. On the basis of these studies, the use of antioxidant supplements by the general population increased substantially and became a multibillion dollar industry. Despite strong biological evidence and promising observational data, more rigorous scientific evaluation did not support a causational relationship between vitamin supplements and lowering coronary artery disease risk. Several prospective, double-blind, placebo-controlled trials showed no benefit and possibly harmful effects. Therapies such as angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and statins, which are known to have benefit in preventing and treating atherosclerosis by reducing blood pressure and cholesterol, also have a "pleiotropic" effect in reducing the formation of reactive oxygen species (ROS). Advances in molecular biology and the study of ROS led to a better understanding of the mechanisms that govern their production and role in atherogenesis. This progress identified unforeseen pathways by which these drugs favorably alter the balance in ROS production, and have raised possibilities for future targeted therapies in the prevention of atherosclerosis.
Collapse
|
44
|
Novel insights into the vasoprotective role of heme oxygenase-1. Int J Hypertens 2012; 2012:127910. [PMID: 22518279 PMCID: PMC3296201 DOI: 10.1155/2012/127910] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/12/2011] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular risk factors contribute to enhanced oxidative stress which leads to endothelial dysfunction. These events trigger platelet activation and their interaction with leukocytes and endothelial cells, thus contributing to the induction of chronic inflammatory processes at the vascular wall and to the development of atherosclerotic lesions and atherothrombosis. In this scenario, endogenous antioxidant pathways are induced to restrain the development of vascular disease. In the present paper, we will discuss the role of heme oxygenase (HO)-1 which is an enzyme of the heme catabolism and cleaves heme to form biliverdin and carbon monoxide (CO). Biliverdin is reduced enzymatically to the potent antioxidant bilirubin. Recent evidence supports the involvement of HO-1 in the antioxidant and antiinflammatory effect of cyclooxygenase(COX)-2-dependent prostacyclin in the vasculature. Moreover, the role of HO-1 in estrogen vasoprotection is emerging. Finally, possible strategies to develop novel therapeutics against cardiovascular disease by targeting the induction of HO-1 will be discussed.
Collapse
|
45
|
Vasculitis, Atherosclerosis, and Altered HDL Composition in Heme-Oxygenase-1-Knockout Mice. Int J Hypertens 2012; 2012:948203. [PMID: 22518297 PMCID: PMC3296294 DOI: 10.1155/2012/948203] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 11/21/2011] [Accepted: 11/29/2011] [Indexed: 11/23/2022] Open
Abstract
To elucidate roles of heme oxygenase-1 (HO-1) in cardiovascular system, we have analyzed one-year-old HO-1-knockout mice. Homozygous HO-1-knockout mice had severe aortitis and coronary arteritis with mononuclear cellular infiltration and fatty streak formation even on a standard chow diet. Levels of plasma total cholesterol and HDL were similar among the three genotypes. However, homozygous HO-1-knockout mice had lower body weight and plasma triglyceride. HO-1-deficiency resulted in alteration of the composition of HDL. The ratio of apolipoprotein AI to AII in HO-1-knockout mice was reduced about 10-fold as compared to wild-type mice. In addition, paraoxonase, an enzyme against oxidative stress, was reduced less than 50% in HO-1-knockout mice. The knockout mice also exhibited significant elevation of plasma lipid hydroperoxides. This study using aged HO-1-knockout mice strengthened the idea that HO-1 functions to suppress systemic inflammation in artery wall and prevents plasma lipid peroxidation.
Collapse
|
46
|
Wu Y, Dong Y, Song P, Zou MH. Activation of the AMP-activated protein kinase (AMPK) by nitrated lipids in endothelial cells. PLoS One 2012; 7:e31056. [PMID: 22363546 PMCID: PMC3281919 DOI: 10.1371/journal.pone.0031056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/31/2011] [Indexed: 02/07/2023] Open
Abstract
The AMP-activated protein kinase (AMPK) is an important regulator of endothelial metabolic and functional homeostasis. Here, we examined the regulation of AMPK by nitrated oleic acid (OA-NO(2)) and investigated the implications in endothelial function. Treatment of bovine aortic endothelial cells (BAECs) with OA-NO(2) induced a significant increase in both AMPK-Thr172 phosphorylation and AMPK activity as well as upregulation of heme oxygenase (HO)-1 and hypoxia-inducible factor (HIF)-1α. Pharmacologic inhibition or genetic ablation of HO-1 or HIF-1α abolished OA-NO(2)-induced AMPK phosphorylation. OA-NO(2) induced a dramatic increase in extracellular signal-regulated kinase (ERK)1/2 phosphorylation that was abrogated by the HO-1 inhibitor, zinc deuteroporphyrin IX 2,4-bis-ethylene glycol (ZnBG). Inhibition of ERK1/2 using UO126 or PD98059 reduced but did not abolish OA-NO(2)-induced HIF-1α upregulation, suggesting that OA-NO(2)/HO-1-initiated HIF-1α induction is partially dependent on ERK1/2 activity. In addition, OA-NO(2) enhanced endothelial intracellular Ca(2+), an effect that was inhibited by the HIF-1α inhibitor, YC-1, and by HIF-1α siRNA. These results implicate the involvement of HIF-1α. Experiments using the Ca(2+)/calmodulin-dependent protein kinase kinase (CaMKK) inhibitor STO-609, the selective CaMKII inhibitor KN-93, and an isoform-specific siRNA demonstrated that OA-NO(2)-induced AMPK phosphorylation was dependent on CaMKKβ. Together, these results demonstrate that OA-NO(2) activates AMPK in endothelial cells via an HO-1-dependent mechanism that increases HIF-1α protein expression and Ca(2+)/CaMKKβ activation.
Collapse
Affiliation(s)
- Yong Wu
- Section of Molecular Medicine, Department of Medicine, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Biochemistry, University of California, Riverside, Riverside, California, United States of America
| | - Yunzhou Dong
- Section of Molecular Medicine, Department of Medicine, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Ping Song
- Section of Molecular Medicine, Department of Medicine, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Ming-Hui Zou
- Section of Molecular Medicine, Department of Medicine, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
47
|
Resveratrol protects rats from Aβ-induced neurotoxicity by the reduction of iNOS expression and lipid peroxidation. PLoS One 2011; 6:e29102. [PMID: 22220203 PMCID: PMC3248406 DOI: 10.1371/journal.pone.0029102] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 11/21/2011] [Indexed: 01/14/2023] Open
Abstract
Alzheimer disease (AD) is an age-dependent neurodegenerative disease characterized by the formation of β–amyloid (Aβ)-containing senile plaque. The disease could be induced by the administration of Aβ peptide, which was also known to upregulate inducible nitric oxide synthase (iNOS) and stimulate neuronal apoptosis. The present study is aimed to elucidate the cellular effect of resveratrol, a natural phytoestrogen with neuroprotective activities, on Aβ-induced hippocampal neuron loss and memory impairment. On adult Sprague-Dawley rats, we found the injection of Aβ could result in a significant impairment in spatial memory, a marked increase in the cellular level of iNOS and lipid peroxidation, and an apparent decrease in the expression of heme oxygenase-1 (HO-1). By combining the treatment with Aβ, resveratrol was able to confer a significant improvement in spatial memory, and protect animals from Aβ-induced neurotoxicity. These neurological protection effects of resveratrol were associated with a reduction in the cellular levels of iNOS and lipid peroxidation and an increase in the production of HO-1. Moreover, the similar neurological and cellular response were also observed when Aβ treatment was combined with the administration of a NOS inhibitor, N(G)-nitro-L-arginine methyl ester hydrochloride (L-NAME). These findings strongly implicate that iNOS is involved in the Aβ-induced lipid peroxidation and HO-1 downregulation, and resveratrol protects animals from Aβ-induced neurotoxicity by suppressing iNOS production.
Collapse
|
48
|
Heme oxygenase-1 inhibits progression and destabilization of vulnerable plaques in a rabbit model of atherosclerosis. Eur J Pharmacol 2011; 672:143-52. [DOI: 10.1016/j.ejphar.2011.09.188] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 09/23/2011] [Accepted: 09/30/2011] [Indexed: 12/29/2022]
|
49
|
Cho HC. The Relationship among Homocysteine, Bilirubin, and Diabetic Retinopathy. Diabetes Metab J 2011; 35:595-601. [PMID: 22247902 PMCID: PMC3253970 DOI: 10.4093/dmj.2011.35.6.595] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 06/07/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diabetic retinopathy is a common microvascular complication of diabetes mellitus (DM) and the leading cause of blindness in adults. Homocysteine, a risk factor with toxic effects on vascular endothelial cells, and bilirubin, a protectant with antioxidant and anti-inflammatory properties on the vasculature, have been reported to be linked to vaso-occlusive disorders. Therefore, the author of the present study investigated the association between the levels of plasma homocysteine and serum total bilirubin and the incidence of diabetic retinopathy as a chronic microvascular complication in patients with type 2 diabetes mellitus (T2DM). METHODS A total of 102 patients with T2DM who visited our hospital from January 2009 to January 2010 were assessed. RESULTS Of the 102 patients, the prevalence of diabetic retinopathy was 67 cases (65.7%) according to clinical ophthalmic examination. The duration of DM (P<0.001), age (P=0.003), fasting blood glucose (P=0.045) and urine albumin-creatinine ratio (P=0.015) in univariate analysis and plasma homocysteine level (P=0.038), duration of DM (P=0.001), and total bilirubin level (P=0.012) in multiple logistic regression analysis were statistically significantly associated with the incidence of diabetic retinopathy. CONCLUSION The present study indicates that homocysteine and bilirubin may be useful biomarkers for increased risk of diabetic retinopathy since retinopathy in patients with T2DM was linked to higher plasma homocysteine level and decreased serum total bilirubin level.
Collapse
Affiliation(s)
- Ho Chan Cho
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, Korea
| |
Collapse
|
50
|
Kadl A, Sharma PR, Chen W, Agrawal R, Meher AK, Rudraiah S, Grubbs N, Sharma R, Leitinger N. Oxidized phospholipid-induced inflammation is mediated by Toll-like receptor 2. Free Radic Biol Med 2011; 51:1903-9. [PMID: 21925592 PMCID: PMC3197756 DOI: 10.1016/j.freeradbiomed.2011.08.026] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/22/2011] [Accepted: 08/23/2011] [Indexed: 12/18/2022]
Abstract
Oxidative tissue damage is a hallmark of many chronic inflammatory diseases. However, the precise mechanisms linking oxidative changes to inflammatory reactions remain unclear. Herein we show that Toll-like receptor 2 (TLR2) translates oxidative tissue damage into inflammatory responses by mediating the effects of oxidized phospholipids. Intraperitoneal injection of oxidized 1-palmitoyl-2-arachidonyl-sn-3-glycerophosphorylcholine (OxPAPC) resulted in upregulation of inflammatory genes in wild-type, but not in TLR2(-/-) mice. In vitro, OxPAPC induced TLR2 (but not TLR4)-dependent inflammatory gene expression and JNK and p38 signaling in macrophages. Induction of TLR2-dependent gene expression required reducible functional groups on sn-2 acyl chains of oxidized phospholipids, as well as serum cofactors. Finally, TLR2(-/-) mice were protected against carbon tetrachloride-induced oxidative tissue damage and inflammation, which was accompanied by accumulation of oxidized phospholipids in livers. Together, our findings demonstrate that TLR2 mediates cellular responses to oxidative tissue damage and they provide new insights into how oxidative stress is linked to acute and chronic inflammation.
Collapse
Affiliation(s)
- Alexandra Kadl
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Poonam R. Sharma
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Wenshu Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rachana Agrawal
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Akshaya K. Meher
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Swetha Rudraiah
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Nathaniel Grubbs
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rahul Sharma
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|