51
|
Endothelial cellular senescence is inhibited by liver X receptor activation with an additional mechanism for its atheroprotection in diabetes. Proc Natl Acad Sci U S A 2014; 111:1168-73. [PMID: 24398515 DOI: 10.1073/pnas.1322153111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Senescence of vascular endothelial cells leads to endothelial dysfunction and contributes to the progression of atherosclerosis. Liver X receptors (LXRs) are nuclear receptors whose activation protects against atherosclerosis by transcriptional regulation of genes important in promoting cholesterol efflux and inhibiting inflammation. Here we found that LXR activation with specific ligands reduced the increase in senescence-associated (SA) β-gal activity, a senescence marker, and reversed the decrease in telomerase activity, a replicative senescence marker, in human endothelial cells under high glucose. This effect of LXR activation was associated with reduced reactive oxygen species and increased endothelial NO synthase activity. A series of experiments that used siRNAs indicated that LXRβ mediates the prevention of endothelial cellular senescence, and that sterol regulatory element binding protein-1, which was up-regulated as a direct LXRβ target gene, may act as a brake of endothelial cellular senescence. Although oral administration of the LXR ligand led to severe fatty liver in diabetic rats, concomitant therapy with metformin avoided the development of hepatic steatosis. However, the preventive effect of the LXR ligand on SA β-gal-stained cells in diabetic aortic endothelium was preserved even if metformin was coadministered. Taken together, our studies demonstrate that an additional mechanism, such as the regulation of endothelial cellular senescence, is related to the antiatherogenic properties of LXRs, and concomitant treatment with metformin may provide a clinically useful therapeutic strategy to alleviate an LXR activation-mediated adverse effects on liver triglyceride metabolism.
Collapse
|
52
|
Zhou HG, Liu L, Zhang Y, Huang YY, Tao YH, Zhang S, Su JJ, Tang YP, Guo ZL, Hu RM, Dong Q. Glutathione prevents free fatty acids-induced oxidative stress and apoptosis in human brain vascular endothelial cells through Akt pathway. CNS Neurosci Ther 2013; 19:252-61. [PMID: 23521913 DOI: 10.1111/cns.12068] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 01/13/2013] [Accepted: 01/14/2013] [Indexed: 12/20/2022] Open
Abstract
AIMS The damage of human brain vascular endothelial cells (HBVECs) is the key pathogenesis of diabetes-associated cerebral vascular complications. The aim of this study was to elucidate the effects of glutathione (GSH) on free fatty acids (FFAs)-induced HBVECs apoptosis, oxidative stress, and the involved possible signaling pathway. METHODS After culturing HBVECs for 72 h with GSH and FFAs, we determined cell proliferation by CCK8, detected apoptosis by caspase-3 and Annexin V-FITC/PI staining, and judged oxygen stress by determining the reactive oxygen species (ROS) and the mitochondrial membrane potential (MMP). We investigated whether the Akt pathway was involved in FFAs-induced signaling pathway alteration and whether GSH influenced the above effects. RESULTS After being cultured in 200 μM FFAs for 72 h, the HBVECs proliferation significantly decreased; HBVECs apoptosis increased; the ROS levels increased; and the HBVECs MMP subsequently decreased. FFAs induced a significant decrease in phosphorylated active Akt. These alterations were obviously prevented when 1 mM GSH was added to culture medium containing FFAs, and the above effects of GSH were blocked by Akt inhibitor. CONCLUSION GSH may prevent FFAs-induced HBVECs damage, oxidative stress, and apoptosis through activating the Akt pathway.
Collapse
Affiliation(s)
- Hou-Guang Zhou
- Department of Geriatrics Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Venkataraman K, Khurana S, Tai TC. Oxidative stress in aging--matters of the heart and mind. Int J Mol Sci 2013; 14:17897-925. [PMID: 24002027 PMCID: PMC3794759 DOI: 10.3390/ijms140917897] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/10/2013] [Accepted: 08/15/2013] [Indexed: 01/15/2023] Open
Abstract
Oxidative damage is considered to be the primary cause of several aging associated disease pathologies. Cumulative oxidative damage tends to be pervasive among cellular macromolecules, impacting proteins, lipids, RNA and DNA of cells. At a systemic level, events subsequent to oxidative damage induce an inflammatory response to sites of oxidative damage, often contributing to additional oxidative stress. At a cellular level, oxidative damage to mitochondria results in acidification of the cytoplasm and release of cytochrome c, causing apoptosis. This review summarizes findings in the literature on oxidative stress and consequent damage on cells and tissues of the cardiovascular system and the central nervous system, with a focus on aging-related diseases that have well-documented evidence of oxidative damage in initiation and/or progression of the disease. The current understanding of the cellular mechanisms with a focus on macromolecular damage, impacted cellular pathways and gross morphological changes associated with oxidative damage is also reviewed. Additionally, the impact of calorific restriction with its profound impact on cardiovascular and neuronal aging is addressed.
Collapse
Affiliation(s)
- Krishnan Venkataraman
- Department of Gerontology, Huntington University, Sudbury, ON P3E 2C6, Canada; E-Mail:
| | - Sandhya Khurana
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; E-Mail:
| | - T. C. Tai
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; E-Mail:
- Department of Biology, Department of Chemistry and Biochemistry, Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-705-662-7239; Fax: +1-705-675-4858
| |
Collapse
|
54
|
Ozone oxidative preconditioning prevents atherosclerosis development in New Zealand White rabbits. J Cardiovasc Pharmacol 2013; 61:160-5. [PMID: 23222311 DOI: 10.1097/fjc.0b013e31827a820d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Atherosclerosis is a major cause of death in the Western World. It is known that Lipofundin 20% induces atherosclerotic lesions, whereas ozone at low doses has been satisfactorily used in the prevention of oxidative stress-associated pathologies, such as coronary artery diseases. The aim of the present work was to evaluate the effects of ozone therapy on Lipofundin-induced atherosclerotic lesions in New Zealand White rabbits. Ozone (1 mg), mixed with oxygen as passive carrier, was administered by rectal insufflation during 15 sessions in 5 weeks. Then, the animals were intravenously treated with 2 mL/kg of Lipofundin, daily during 8 days. Animals were euthanized and eosin and hematoxylin staining was used for aortic histopathological analysis. The biomarkers of oxidative stress and lipid profile in serum were determined by spectrophotometric techniques. The results demonstrated that ozone induced inhibitory effects on aortic lesions formation. On the other hand, a reduction of biomolecular damage and an increase of antioxidant systems were observed at the end of the experiment. The serum lipids profiles were not modified after only 1 cycle of ozone treatment. Our results reinforced the hypotheses that antioxidant effects induced by ozone in the context of atherosclerosis demonstrate the antiatherogenic properties of the gas in the experimental conditions of this study.
Collapse
|
55
|
Silicic acid in drinking water prevents age-related alterations in the endothelium-dependent vascular relaxation modulating eNOS and AQP1 expression in experimental mice: an immunohistochemical study. Acta Histochem 2013. [PMID: 23177919 DOI: 10.1016/j.acthis.2012.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The maintenance of endothelial integrity is of great importance in coping with age-related vascular alterations. Endothelium-derived nitric oxide is one of the various vasoactive substances able to regulate vascular tone and homeostasis, and whose decrease is known to be related with senescence in endothelial cells. There are reports on the efficacy of silicon, especially as silicic acid, in protecting vascular integrity during age-related vascular diseases. The aim of this study was to evaluate the ability of supplementation of silicic acid in drinking water in the maintenance of vascular health in a mouse model of early physiological aging. In particular, we evaluated the relationship between Si supplementation and endothelial nitric oxide synthase (eNOS) expression, taking into account also the aquaporin-1 (AQP-1) isoform that, as recently reported, seems to be involved in nitric oxide transport across cell membranes. Our results showed that silicic acid supplementation increased both eNOS and AQP-1 expression, suggesting that silicic acid modulation of endothelial nitric oxide synthase and aquaporin-1 could represent a potential strategy against age-related vascular senescence.
Collapse
|
56
|
Rogers SC, Zhang X, Azhar G, Luo S, Wei JY. Exposure to high or low glucose levels accelerates the appearance of markers of endothelial cell senescence and induces dysregulation of nitric oxide synthase. J Gerontol A Biol Sci Med Sci 2013; 68:1469-81. [PMID: 23585419 DOI: 10.1093/gerona/glt033] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To test the hypothesis that aging impairs endothelial cell response to glucose stress, we utilized a human umbilical vein endothelial cell in vitro model in which clinically relevant concentrations of normal (5.5 mM), high (25 mM), and low (1.5mM) glucose were tested. With advancing population doubling, exposure to normal glucose gradually decreased endothelial nitric oxide synthase expression and activity, resulting in slow, progressive development of markers of cell senescence (by population doubling level [PDL] 44). High or low glucose treatment accelerated the appearance of markers of senescence (by ~PDL 35) along with declines in endothelial nitric oxide synthase expression and activity. Human umbilical vein endothelial cells exposed to alternating low and high glucose gave even more rapid acceleration in the appearance of markers of senescence (by ~PDL 18) and reduction in endothelial nitric oxide synthase levels. Thus, exposure to low and high glucose induces earlier appearance of markers of endothelial cell senescence and dysregulation of the nitric oxide synthase gene and protein expression and function. These findings will help to elucidate endothelial dysfunction associated with glucose intolerance and improve future therapy for diabetic seniors.
Collapse
Affiliation(s)
- Steven C Rogers
- Reynolds Institute on Aging, University of Arkansas for Medical Sciences, 629 Jack Stephens Drive, Little Rock, AR 72205.
| | | | | | | | | |
Collapse
|
57
|
Xiao-Hong D, Chang-Qin X, Jian-Hua H, Wen-Jiang Z, Bing S. Icariin delays homocysteine-induced endothelial cellular senescence involving activation of the PI3K/AKT-eNOS signaling pathway. PHARMACEUTICAL BIOLOGY 2013; 51:433-440. [PMID: 23336586 DOI: 10.3109/13880209.2012.738332] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
CONTEXT Homocysteine-induced endothelial cellular senescence may contribute to some cardiovascular disorders. Icariin (ICA), a flavonoid derived from Epimedium sagittatum Maxim. (Berberidaceae), has been reported to increase production of nitric oxide (NO) and reduce reactive oxygen species (ROS) levels in human umbilical vein endothelial cells (HUVECs). OBJECTIVE To observe the effects of ICA on homocysteine-induced senescence and the underlying mechanisms in HUVECs. MATERIALS AND METHODS ICA at concentrations of 0.1, 1, and 5 μM was added into homocysteine pretreated HUVECs. Cellular senescence was assayed by senescence-associated β-galactosidase (SA-β-gal) staining and cumulative population doublings (CPDs). ICA (5 μM) was given orally to homocysteine-treated rats, luminal surface of aortic artery of rats was subjected to SA-β-gal staining. Protein expression was measured by western blot. RESULTS Homocysteine significantly increased cellular senescence both in vitro and in vivo. After treatment by ICA, the percentage of SA-β-gal-positive cells, and the ROS level significantly decreased. The CPDs were partially restored. ICA also significantly reduced the mean density of SA-β-gal staining in vivo. We found that NO production and phosphorylation of AKT, ERK, and endothelial NO synthase (eNOS) were elevated by ICA in HUVECs. Furthermore, the increased level of NO production was fully abolished by the phosphatidylinositol-3-kinase (PI3K) inhibitor wortmannin. The mitogen-activated protein kinase (MEK) inhibitor PD98059, which can inhibit phosphorylation of ERK, did not show this ability. DISCUSSION AND CONCLUSION Our results indicate that ICA delays homocyteine-induced endothelial senescence in vitro and in vivo. Activation of PI3K/Akt-eNOS-dependent signaling pathway may be responsible for this efficacy of ICA.
Collapse
Affiliation(s)
- Duan Xiao-Hong
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
58
|
Ghriallais RN, McNamara L, Bruzzi M. Comparison of in vitro human endothelial cell response to self-expanding stent deployment in a straight and curved peripheral artery simulator. J R Soc Interface 2013; 10:20120965. [PMID: 23365191 DOI: 10.1098/rsif.2012.0965] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Haemodynamic forces have a synergistic effect on endothelial cell (EC) morphology, proliferation, differentiation and biochemical expression profiles. Alterations to haemodynamic force levels have been observed at curved regions and bifurcations of arteries but also around struts of stented arteries, and are also known to be associated with various vascular pathologies. Therefore, curvature in combination with stenting might create a pro-atherosclerotic environment compared with stenting in a straight vessel, but this has never been investigated. The goal of this study was to compare EC morphology, proliferation and differentiation within in vitro models of curved stented peripheral vessel models with those of straight and unstented vessels. These models were generated using both static conditions and also subjected to 24 h of stimulation in a peripheral artery bioreactor. Medical-grade silicone tubes were seeded with human umbilical vein endothelial cells to produce pseudovessels that were then stented and subjected to 24 h of physiological levels of pulsatile pressure, radial distention and shear stress. Changes in cell number, orientation and nitric oxide (NO) production were assessed in straight, curved, non-stented and stented pseudovessels. We report that curved pseudovessels lead to higher EC numbers with random orientation and lower NO production per cell compared with straight pseudovessels after 24 h of biomechanical stimulation. Both stented curved and stented straight pseudovessels had lower NO production per cell than corresponding unstented pseudovessels. However, in contrast to straight stented pseudovessels, curved stented pseudovessels had fewer viable cells. The results of this study show, for the first time, that the response of the vascular endothelium is dependent on both curvature and stenting combined, and highlight the necessity for future investigations of the effects of curvature in combination with stenting to fully understand effects on the endothelial layer.
Collapse
Affiliation(s)
- Ríona Ní Ghriallais
- Department of Mechanical and Biomedical Engineering, National University of Ireland, Galway, Ireland.
| | | | | |
Collapse
|
59
|
Abstract
Hypertension is one of the common diseases in the elderly. The prevalence of hypertension markedly increases with advancing age. Both aging and hypertension have a critical role in cardiovascular and cerebrovascular complications. Although aging and hypertension, either independently or collectively, impair endothelial function, aging and hypertension may have similar cascades for the pathogenesis and development of endothelial dysfunction. Nitric oxide (NO) has an important role in regulation of vascular tone. Decrease in NO bioavailability by endothelial dysfunction would lead to elevation of blood pressure. An imbalance of reduced production of NO or increased production of reactive oxygen species, mainly superoxide, may promote endothelial dysfunction. One possible mechanism by which the prevalence of hypertension is increased in relation to aging may be advancing endothelial dysfunction associated with aging through an increase in oxidative stress. In addition, endothelial cell senescence is also involved in aging-related endothelial dysfunction. In this review, we focus on recent findings and interactions between endothelial function, oxidative stress and hypertension in aging.
Collapse
Affiliation(s)
- Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan.
| | | | | |
Collapse
|
60
|
β2-Glycoprotein I inhibits endothelial cell migration through the nuclear factor κB signalling pathway and endothelial nitric oxide synthase activation. Biochem J 2012; 445:125-33. [DOI: 10.1042/bj20111383] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
β2-GPI (β2-glycoprotein I) is a plasma glycoprotein ascribed with an anti-angiogenic function; however, the biological role and molecular basis of its action in cell migration remain unknown. The aim of the present study was to assess the contribution of β2-GPI to HAEC (human aortic endothelial cell) migration and the details of its underlying mechanism. Using wound healing and Boyden chamber assays, we found that β2-GPI inhibited endothelial cell migration, which was restored by its neutralizing antibody. NF-κB (nuclear factor κB) inhibitors and lentiviral siRNA (small interfering RNA) silencing of NF-κB significantly attenuated the inhibitory effect of β2-GPI on cell migration. Moreover, β2-GPI was found to induce IκBα (inhibitor of NF-κB) phosphorylation and translocation of p65 and p50. We further demonstrated that mRNA and protein levels of eNOS [endothelial NO (nitric oxide) synthase] and NO production were all increased by β2-GPI and these effects were remarkably inhibited by NF-κB inhibitors and siRNAs of p65 and p50. Furthermore, β2-GPI-mediated inhibition of cell migration was reversed by eNOS inhibitors and eNOS siRNAs. The findings of the present study provide novel insight into the ability of β2-GPI to inhibit endothelial cell migration predominantly through the NF-κB/eNOS/NO signalling pathway, which indicates a potential direction for clinical therapy in vascular diseases.
Collapse
|
61
|
Novella S, Dantas AP, Segarra G, Medina P, Hermenegildo C. Vascular Aging in Women: is Estrogen the Fountain of Youth? Front Physiol 2012; 3:165. [PMID: 22685434 PMCID: PMC3368545 DOI: 10.3389/fphys.2012.00165] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/08/2012] [Indexed: 11/13/2022] Open
Abstract
Aging is associated with structural and functional changes in the vasculature, including endothelial dysfunction, arterial stiffening and remodeling, impaired angiogenesis, and defective vascular repair, and with increased prevalence of atherosclerosis. Cardiovascular risk is similar for older men and women, but lower in women during their fertile years. This age- and sex-related difference points to estrogen as a protective factor because menopause is marked by the loss of endogenous estrogen production. Experimental and some clinical studies have attributed most of the protective effects of estrogen to its modulatory action on vascular endothelium. Estrogen promotes endothelial-derived NO production through increased expression and activity of endothelial nitric oxide synthase, and modulates prostacyclin and thromboxane A(2) release. The thromboxane A(2) pathway is key to regulating vascular tone in females. Despite all the experimental evidence, some clinical trials have reported no cardiovascular benefit from estrogen replacement therapy in older postmenopausal women. The "Timing Hypothesis," which states that estrogen-mediated vascular benefits occur only before the detrimental effects of aging are established in the vasculature, offers a possible explanation for these discrepancies. Nevertheless, a gap remains in current knowledge of cardiovascular aging mechanisms in women. This review comprises clinical and experimental data on the effects of aging, estrogens, and hormone replacement therapy on vascular function of females. We aim to clarify how menopause and aging contribute jointly to vascular aging and how estrogen modulates vascular response at different ages.
Collapse
Affiliation(s)
- Susana Novella
- Departamento de Fisiología, Universitat de València Valencia, Spain
| | | | | | | | | |
Collapse
|
62
|
El Assar M, Angulo J, Vallejo S, Peiró C, Sánchez-Ferrer CF, Rodríguez-Mañas L. Mechanisms involved in the aging-induced vascular dysfunction. Front Physiol 2012; 3:132. [PMID: 22783194 PMCID: PMC3361078 DOI: 10.3389/fphys.2012.00132] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 04/20/2012] [Indexed: 12/25/2022] Open
Abstract
Vascular aging is a key process determining health status of aged population. Aging is an independent cardiovascular risk factor associated to an impairment of endothelial function, which is a very early and important event leading to cardiovascular disease. Vascular aging, formerly being considered an immutable and inexorable risk factor, is now viewed as a target process for intervention in order to achieve a healthier old age. A further knowledge of the mechanisms underlying the age-related vascular dysfunction is required to design an adequate therapeutic strategy to prevent or restore this impairment of vascular functionality. Among the proposed mechanisms that contribute to age-dependent endothelial dysfunction, this review is focused on the following aspects occurring into the vascular wall: (1) the reduction of nitric oxide (NO) bioavailability, caused by diminished NO synthesis and/or by augmented NO scavenging due to oxidative stress, leading to peroxynitrite formation (ONOO(-)); (2) the possible sources involved in the enhancement of oxidative stress; (3) the increased activity of vasoconstrictor factors; and (4) the development of a low-grade pro-inflammatory environment. Synergisms and interactions between all these pathways are also analyzed. Finally, a brief summary of some cellular mechanisms related to endothelial cell senescence (including telomere and telomerase, stress-induced senescence, as well as sirtuins) are implemented, as they are likely involved in the age-dependent endothelial dysfunction, as well as in the lower vascular repairing capacity observed in the elderly. Prevention or reversion of those mechanisms leading to endothelial dysfunction through life style modifications or pharmacological interventions could markedly improve cardiovascular health in older people.
Collapse
Affiliation(s)
- Mariam El Assar
- Fundación para la Investigación Biomédica, Hospital Universitario de Getafe Madrid, Spain
| | | | | | | | | | | |
Collapse
|
63
|
Jane-Wit D, Chun HJ. Mechanisms of dysfunction in senescent pulmonary endothelium. J Gerontol A Biol Sci Med Sci 2012; 67:236-41. [PMID: 22396472 DOI: 10.1093/gerona/glr248] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Age-dependent changes in pulmonary endothelium contribute to worsened clinical outcomes in elderly individuals. Due to altered pulmonary endothelial responses, older participants have increased vulnerability to infection-related sequelae, higher prevalence of pulmonary hypertension, mitigated DNA repair mechanisms, and attenuated parenchymal healing. Aberrant signaling in pulmonary endothelium undergird these clinical processes. In this review, we provide an overview of the work that has elucidated age-related molecular derangements in pulmonary endothelial cells. In particular, we summarize studies describing mishandling of intracellular reactive oxygen species, pathological nitric oxide signaling, and deficient recruitment of endothelial stem cell precursors. We conclude with a summary of potential future avenues of investigation. The signaling pathways associated with pulmonary endothelial senescence reviewed herein suggest a number of putative therapeutic drug targets. Further elucidation of the cellular processes associated with aging in the pulmonary endothelium may provide critical insights into the rational design of therapies that may subvert or even reverse the effects of aging on a molecular level.
Collapse
Affiliation(s)
- Daniel Jane-Wit
- Section of Cardiovascular Medicine, Yale University School of Medicine, 300 George St, Room 773K, New Haven, CT 06511, USA
| | | |
Collapse
|
64
|
Delgado Roche L, Acosta Medina E, Fraga Pérez Á, Bécquer Viart MA, Soto López Y, Falcón Cama V, Vázquez López AM, Martínez-Sánchez G, Fernández-Sánchez E. Lipofundin-induced hyperlipidemia promotes oxidative stress and atherosclerotic lesions in new zealand white rabbits. Int J Vasc Med 2011; 2012:898769. [PMID: 21977325 PMCID: PMC3184413 DOI: 10.1155/2012/898769] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 06/21/2011] [Accepted: 07/23/2011] [Indexed: 01/21/2023] Open
Abstract
Atherosclerosis represents a major cause of death in the world. It is known that Lipofundin 20% induces atherosclerotic lesions in rabbits, but its effects on serum lipids behaviour and redox environment have not been addressed. In this study, New Zealand rabbits were treated with 2 mL/kg of Lipofundin for 8 days. Then, redox biomarkers and serum lipids were determined spectrophotometrically. On the other hand, the development of atherosclerotic lesions was confirmed by eosin/hematoxylin staining and electron microscopy. At the end of the experiment, total cholesterol, triglycerides, cholesterol-LDL, and cholesterol-HDL levels were significantly increased. Also, a high index of biomolecules damage, a disruption of both enzymatic and nonenzymatic defenses, and a reduction of nitric oxide were observed. Our data demonstrated that Lipofundin 20% induces hyperlipidemia, which promotes an oxidative stress state. Due to the importance of these phenomena as risk factors for atherogenesis, we suggest that Lipofundin induces atherosclerosis mainly through these mechanisms.
Collapse
Affiliation(s)
- Livan Delgado Roche
- Center of Studies for Research and Biological Evaluations, Pharmacy and Food Science College, University of Havana, PO. Box 13 600, La Coronela, La Lisa, Havana 13600, Cuba
| | - Emilio Acosta Medina
- Center of Studies for Research and Biological Evaluations, Pharmacy and Food Science College, University of Havana, PO. Box 13 600, La Coronela, La Lisa, Havana 13600, Cuba
| | - Ángela Fraga Pérez
- Center of Studies for Research and Biological Evaluations, Pharmacy and Food Science College, University of Havana, PO. Box 13 600, La Coronela, La Lisa, Havana 13600, Cuba
| | - María A. Bécquer Viart
- Center of Studies for Research and Biological Evaluations, Pharmacy and Food Science College, University of Havana, PO. Box 13 600, La Coronela, La Lisa, Havana 13600, Cuba
| | - Yosdel Soto López
- Department of Antibody Engineering, Center of Molecular Immunology, Havana 11600, Cuba
| | - Viviana Falcón Cama
- Department of Electron Microscopy, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba
| | - Ana M. Vázquez López
- Department of Antibody Engineering, Center of Molecular Immunology, Havana 11600, Cuba
| | | | - Eduardo Fernández-Sánchez
- Center of Studies for Research and Biological Evaluations, Pharmacy and Food Science College, University of Havana, PO. Box 13 600, La Coronela, La Lisa, Havana 13600, Cuba
| |
Collapse
|
65
|
Role of microRNAs in endothelial inflammation and senescence. Mol Biol Rep 2011; 39:4509-18. [PMID: 21952822 DOI: 10.1007/s11033-011-1241-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 09/14/2011] [Indexed: 01/07/2023]
Abstract
The functionality of endothelial cells is fundamental for the homoeostasis of the vascular system. Increasing evidence shows that endothelial inflammation and senescence contribute greatly to multiple vascular diseases including atherosclerosis. However, little is known regarding the complex upstream regulators of gene expression and translation involved in these responses. MicroRNAs (miRNAs) have emerged as a novel class of endogenous, small, non-coding RNAs that negatively regulate over 30% of genes in a cell via degradation or translational inhibition of their target mRNAs. During the past few years, miRNAs have emerged as key regulators for endothelial biology and function. Endothelial inflammation is critically regulated by miRNAs such as miR-126 and miR-10a in vitro and in vivo. Endothelial aging is additionally controlled by miR-217 and miR-34a. In this review, we summarize the role of miRNAs and their target genes in endothelial inflammation and senescence, and discuss their applicability as drug targets.
Collapse
|
66
|
Cao D, Li H, Yi J, Zhang J, Che H, Cao J, Yang L, Zhu C, Jiang W. Antioxidant properties of the mung bean flavonoids on alleviating heat stress. PLoS One 2011; 6:e21071. [PMID: 21695166 PMCID: PMC3112222 DOI: 10.1371/journal.pone.0021071] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 05/18/2011] [Indexed: 01/29/2023] Open
Abstract
Background It is a widespread belief in Asian countries that mung bean soup (MBS) may afford a protective effect against heat stress. Lack of evidence supports MBS conferring a benefit in addition to water. Results Here we show that vitexin and isovitexin are the major antioxidant components in mungbean (more than 96% of them existing in the bean seed coat), and both of them could be absorbed via gavage into rat plasma. In the plasma of rats fed with mungbean coat extract before or after exposure to heat stress, the levels of malonaldehyde and activities of lactate dehydrogenase and nitric oxide synthase were remarkably reduced; the levels of total antioxidant capacity and glutathione (a quantitative assessment of oxidative stress) were significantly enhanced. Conclusions Our results demonstrate that MBS can play additional roles to prevent heat stress injury. Characterization of the mechanisms underlying mungbean beneficial effects should help in the design of diet therapy strategies to alleviate heat stress, as well as provide reference for searching natural medicines against oxidative stress induced diseases.
Collapse
Affiliation(s)
- Dongdong Cao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - He Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Jianyong Yi
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Jingjing Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Huilian Che
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Jiankang Cao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Liu Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Chunqiu Zhu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Weibo Jiang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
67
|
Matsui-Hirai H, Hayashi T, Yamamoto S, Ina K, Maeda M, Kotani H, Iguchi A, Ignarro LJ, Hattori Y. Dose-dependent modulatory effects of insulin on glucose-induced endothelial senescence in vitro and in vivo: a relationship between telomeres and nitric oxide. J Pharmacol Exp Ther 2011; 337:591-9. [PMID: 21357660 DOI: 10.1124/jpet.110.177584] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The elderly are prone to postprandial hyperglycemia that increases their cardiovascular risk. Although insulin therapy is necessary to treat diabetes, high plasma concentrations of insulin may cause the development of atherosclerosis and accelerate endothelial senescence. We assumed that high glucose causes stress-induced premature senescence and replicative senescence and examined the regulatory role of insulin in endothelial senescence and functions under different glucose conditions. Exposure of human endothelial cells to high glucose (22 mM) for 3 days increased senescence-associated-β-galactosidase activity, a senescence marker, and decreased telomerase activity, a replicative senescence marker. Physiological concentrations of insulin preserved telomere length and delayed endothelial senescence under high-glucose conditions. The effect of insulin under high-glucose conditions was associated with reduced reactive oxygen species and increased nitric oxide (NO). Small interfering RNA targeting endothelial NO synthase reduced the antisenescence effects of insulin. Physiological concentrations of insulin also reversed high glucose-induced increases in p53 and vascular cell adhesion molecule-1 and decreases in senescence marker protein-30. On the other hand, when insulin was given at any concentrations under normal glucose or at high concentrations under high glucose, its ability to promote cellular senescence was unrelated to endothelial NO. Finally, streptozotocin-induced diabetes showed more senescent cells in the aortic endothelium of aged rats compared with age-matched control and insulin-treated animals. Conclusively, the regulatory effects of insulin on endothelial senescence were modulated by the glucose environment. These data may help explain insulin's complicated roles in atherosclerosis in the elderly.
Collapse
Affiliation(s)
- Hisako Matsui-Hirai
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Illi B, Colussi C, Rosati J, Spallotta F, Nanni S, Farsetti A, Capogrossi MC, Gaetano C. NO points to epigenetics in vascular development. Cardiovasc Res 2011; 90:447-56. [PMID: 21345806 DOI: 10.1093/cvr/cvr056] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Our understanding of epigenetic mechanisms important for embryonic vascular development and cardiovascular differentiation is still in its infancy. Although molecular analyses, including massive genome sequencing and/or in vitro/in vivo targeting of specific gene sets, has led to the identification of multiple factors involved in stemness maintenance or in the early processes of embryonic layers specification, very little is known about the epigenetic commitment to cardiovascular lineages. The object of this review will be to outline the state of the art in this field and trace the perspective therapeutic consequences of studies aimed at elucidating fundamental epigenetic networks. Special attention will be paid to the emerging role of nitric oxide in this field.
Collapse
Affiliation(s)
- Barbara Illi
- Mendel Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Cardenas JC, Owens AP, Krishnamurthy J, Sharpless NE, Whinna HC, Church FC. Overexpression of the cell cycle inhibitor p16INK4a promotes a prothrombotic phenotype following vascular injury in mice. Arterioscler Thromb Vasc Biol 2011; 31:827-33. [PMID: 21233453 DOI: 10.1161/atvbaha.110.221721] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Age-associated cellular senescence is thought to promote vascular dysfunction. p16(INK4a) is a cell cycle inhibitor that promotes senescence and is upregulated during normal aging. In this study, we examine the contribution of p16(INK4a) overexpression to venous thrombosis. METHODS AND RESULTS Mice overexpressing p16(INK4a) were studied with 4 different vascular injury models: (1) ferric chloride (FeCl(3)) and (2) Rose Bengal to induce saphenous vein thrombus formation; (3) FeCl(3) and vascular ligation to examine thrombus resolution; and (4) lipopolysaccharide administration to initiate inflammation-induced vascular dysfunction. p16(INK4a) transgenic mice had accelerated occlusion times (13.1 ± 0.4 minutes) compared with normal controls (19.7 ± 1.1 minutes) in the FeCl(3) model and 12.7 ± 2.0 and 18.6 ± 1.9 minutes, respectively in the Rose Bengal model. Moreover, overexpression of p16(INK4a) delayed thrombus resolution compared with normal controls. In response to lipopolysaccharide treatment, the p16(INK4a) transgenic mice showed enhanced thrombin generation in plasma-based calibrated automated thrombography assays. Finally, bone marrow transplantation studies suggested increased p16(INK4a) expression in hematopoietic cells contributes to thrombosis, demonstrating a role for p16(INK4a) expression in venous thrombosis. CONCLUSIONS Venous thrombosis is augmented by overexpression of the cellular senescence protein p16(INK4a).
Collapse
Affiliation(s)
- Jessica C Cardenas
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, NC 27599-7035, USA
| | | | | | | | | | | |
Collapse
|
70
|
Hayashi T. [Senescence and endothelial dysfunction/atherosclerosis]. Nihon Ronen Igakkai Zasshi 2011; 48:142-145. [PMID: 21778629 DOI: 10.3143/geriatrics.48.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
|
71
|
Vassilopoulos SI, Tosios KI, Panis VG, Vrotsos JA. Endothelial cells of oral pyogenic granulomas express eNOS and CD105/endoglin: an immunohistochemical study. J Oral Pathol Med 2010; 40:345-51. [DOI: 10.1111/j.1600-0714.2010.00969.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
72
|
Abstract
Cellular senescence is a specialized form of growth arrest, confined to mitotic cells, induced by various stressful stimuli and characterized by a permanent growth arrest, resistance to apoptosis, an altered pattern of gene expression and the expression of some markers that are characteristic, although not exclusive, to the senescent state. Senescent cells profoundly modify neighboring and remote cells through the production of an altered secretome, eventually leading to inflammation, fibrosis and possibly growth of neoplastic cells. Mammalian aging has been defined as a reduction in the capacity to adequately maintain tissue homeostasis or to repair tissues after injury. Tissue homeostasis and regenerative capacity are nowadays considered to be related to the stem cell pool present in every tissue. For this reason, pathological and patho-physiological conditions characterized by altered tissue homeostasis and impaired regenerative capacity can be viewed as a consequence of the reduction in stem cell number and/or function. Last, cellular senescence is a double-edged sword, since it may inhibit the growth of transformed cells, preventing the occurrence of cancer, while it may facilitate growth of preneoplastic lesions in a paracrine fashion; therefore, interventions targeting this cell response to stress may have a profound impact on many age-related pathologies, ranging from cardiovascular disease to oncology. Aim of this review is to discuss both molecular mechanisms associated with stem cell senescence and interventions that may attenuate or reverse this process.
Collapse
|
73
|
Paradise WA, Vesper BJ, Goel A, Waltonen JD, Altman KW, Haines GK, Radosevich JA. Nitric oxide: perspectives and emerging studies of a well known cytotoxin. Int J Mol Sci 2010; 11:2715-45. [PMID: 20717533 PMCID: PMC2920563 DOI: 10.3390/ijms11072715] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 06/17/2010] [Accepted: 07/13/2010] [Indexed: 12/19/2022] Open
Abstract
The free radical nitric oxide (NO•) is known to play a dual role in human physiology and pathophysiology. At low levels, NO• can protect cells; however, at higher levels, NO• is a known cytotoxin, having been implicated in tumor angiogenesis and progression. While the majority of research devoted to understanding the role of NO• in cancer has to date been tissue-specific, we herein review underlying commonalities of NO• which may well exist among tumors arising from a variety of different sites. We also discuss the role of NO• in human physiology and pathophysiology, including the very important relationship between NO• and the glutathione-transferases, a class of protective enzymes involved in cellular protection. The emerging role of NO• in three main areas of epigenetics—DNA methylation, microRNAs, and histone modifications—is then discussed. Finally, we describe the recent development of a model cell line system in which human tumor cell lines were adapted to high NO• (HNO) levels. We anticipate that these HNO cell lines will serve as a useful tool in the ongoing efforts to better understand the role of NO• in cancer.
Collapse
Affiliation(s)
- William A. Paradise
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
| | - Benjamin J. Vesper
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
| | - Ajay Goel
- Division of Gastroenterology, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, Dallas, TX 75246, USA; E-Mail:
| | - Joshua D. Waltonen
- Department of Otolaryngology, Wake Forest University, Winston-Salem, NC 27157, USA; E-Mail:
| | | | - G. Kenneth Haines
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; E-Mail:
| | - James A. Radosevich
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
- Author to whom correspondence should be addressed; E-Mail:
| |
Collapse
|
74
|
Spallotta F, Rosati J, Straino S, Nanni S, Grasselli A, Ambrosino V, Rotili D, Valente S, Farsetti A, Mai A, Capogrossi MC, Gaetano C, Illi B. Nitric oxide determines mesodermic differentiation of mouse embryonic stem cells by activating class IIa histone deacetylases: potential therapeutic implications in a mouse model of hindlimb ischemia. Stem Cells 2010; 28:431-42. [PMID: 20073046 DOI: 10.1002/stem.300] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In human endothelial cells, nitric oxide (NO) results in class IIa histone deacetylases (HDACs) activation and marked histone deacetylation. It is unknown whether similar epigenetic events occur in embryonic stem cells (ESC) exposed to NO and how this treatment could influence ESC therapeutic potential during tissue regeneration.This study reports that the NO-dependent class IIa HDACs subcellular localization and activity decreases the global acetylation level of H3 histones in ESC and that this phenomenon is associated with the inhibition of Oct4, Nanog, and KLF4 expression. Further, a NO-induced formation of macromolecular complexes including HDAC3, 4, 7, and protein phosphatase 2A (PP2A) have been detected. These processes correlated with the expression of the mesodermal-specific protein brachyury (Bry) and the appearance of several vascular and skeletal muscle differentiation markers. These events were abolished by the class IIa-specific inhibitor MC1568 and by HDAC4 or HDAC7 short interfering RNA (siRNA). The ability of NO to induce mesodermic/cardiovascular gene expression prompted us to evaluate the regenerative potential of these cells in a mouse model of hindlimb ischemia. We found that NO-treated ESCs injected into the cardiac left ventricle selectively localized in the ischemic hindlimb and contributed to the regeneration of muscular and vascular structures. These findings establish a key role for NO and class IIa HDACs modulation in ESC mesodermal commitment and enhanced regenerative potential in vivo.
Collapse
Affiliation(s)
- Francesco Spallotta
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell' Immacolata - Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
The endothelium is vital to the proper functioning in the heart, in particular due to its production of nitric oxide (NO) which regulates vascular tone. Damage to the endothelium contributes to the development of atherosclerosis, and hence to possible myocardial infarction and subsequent heart failure. Like most cells, endothelial cells contain mitochondria, despite their having relatively little dependence on oxidative phosphorylation for ATP production. However, endothelial mitochondria are centrally involved in maintaining the fine regulatory balance between mitochondrial calcium concentration, reactive oxygen species (ROS) production, and NO. This raises the question of whether damage to endothelial mitochondria would have repercussions in terms of the development of heart disease. In fact, increasingly nuanced techniques enabling restricted transgenic expression of antioxidant proteins in mice has demonstrated that mitochondrial ROS do contribute to endothelial damage. New pharmaceutical approaches designed to target protective molecules such as ROS scavengers to the mitochondria promise to be effective in preventing heart disease. As well as protecting cardiomyocytes, these drugs may have the added benefit of preventing damage to the endothelial mitochondria. However, much remains to be done in understanding the contribution that mitochondria make to endothelial function.
Collapse
Affiliation(s)
- Sean Michael Davidson
- Department of Medicine, The Hatter Cardiovascular Institute, University College London Hospital, London WC1E 6HX, UK.
| |
Collapse
|
76
|
Herrera MD, Mingorance C, Rodríguez-Rodríguez R, Alvarez de Sotomayor M. Endothelial dysfunction and aging: an update. Ageing Res Rev 2010; 9:142-52. [PMID: 19619671 DOI: 10.1016/j.arr.2009.07.002] [Citation(s) in RCA: 231] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 12/19/2022]
Abstract
Aging is an important risk factor for the development of many cardiovascular diseases as atherosclerosis and hypertension with a common underlying circumstance: the progressive decline of endothelial function. Vascular endothelial dysfunction occurs during the human aging process and is accompanied by deterioration in the balance between vasodilator and vasoconstriction substances produced by the endothelium. This imbalance is mainly characterized by a progressive reduction of the bioavailability of nitric oxide (NO) and an increase in the production of cyclooxygenase (COX)-derived vasoconstrictor factors. Both circumstances are in turn related to an increased production of reactive oxygen and nitrogen species. The aim of this review is to describe the pathophysiological mechanisms involved in the endothelial function declination that accompanies the multifactorial aging process, including alterations related to oxidative stress and pro-inflammatory cytokines, senescence of endothelial cells and genetic factors.
Collapse
Affiliation(s)
- María Dolores Herrera
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, C/Profesor García González 2, 41012 Seville, Spain.
| | | | | | | |
Collapse
|
77
|
Takahashi S, Nakashima Y, Toda KI. Carnosine facilitates nitric oxide production in endothelial f-2 cells. Biol Pharm Bull 2010; 32:1836-9. [PMID: 19881293 DOI: 10.1248/bpb.32.1836] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined the effect of carnosine (beta-alanyl-histidine) on nitric oxide (NO) production and endothelial NO synthase (eNOS) activation in endothelial F-2 cells. Carnosine enhanced NO production in a dose-dependent manner, and the stimulatory effect of carnosine was observed at concentrations exceeding 5 mM. The carnosine-stimulated NO production was inhibited by N(G)-nitro-L-arginine methyl ester, but not by N(G)-nitro-D-arginine methyl ester. In contrast, beta-alanine, histidine (carnosine components) and anserine (N-methyl carnosine) failed to increase NO production. Carnosine had no effect on NO production for the initial 5 min, but thereafter resulted in a gradual increase in NO production up to 15 min. Carnosine did not induce phosphorylation of eNOS at Ser1177. The carnosine-induced increase in NO production was observed even when extracellular Ca2+ was depleted by ethylene glycol bis(2-aminoethyl ether)-N,N,N'-N'-tetraacetic acid however, the effect was abolished upon depletion of intracellular Ca2+ by BAPTA. After F-2 cells were incubated with carnosine for 4 min, intracellular Ca2+ concentration gradually increased. The carnosine-induced increase in intracellular Ca2+ concentration occurred even in the absence of extracellular Ca2+. These results indicate that carnosine facilitates NO production in endothelial F-2 cells. It is also suggested that eNOS is activated by Ca2+, which might be released from intracellular Ca2+ stores in response to carnosine.
Collapse
Affiliation(s)
- Satoru Takahashi
- First Department of Biochemistry, School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, Yoshino, Nobeoka, Miyazaki 882-0072, Japan.
| | | | | |
Collapse
|
78
|
Gruber HE, Hoelscher GL, Ingram JA, Zinchenko N, Hanley EN. Senescent vs. non-senescent cells in the human annulus in vivo: cell harvest with laser capture microdissection and gene expression studies with microarray analysis. BMC Biotechnol 2010; 10:5. [PMID: 20109216 PMCID: PMC2828399 DOI: 10.1186/1472-6750-10-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 01/28/2010] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Senescent cells are well-recognized in the aging/degenerating human disc. Senescent cells are viable, cannot divide, remain metabolically active and accumulate within the disc over time. Molecular analysis of senescent cells in tissue offers a special challenge since there are no cell surface markers for senescence which would let one use fluorescence-activated cell sorting as a method for separating out senescent cells. METHODS We employed a novel laser capture microdissection (LCM) design to selectively harvest senescent and non-senescent annulus cells in paraffin-embedded tissue, and compared their gene expression with microarray analysis. LCM was used to separately harvest senescent and non-senescent cells from 11 human annulus specimens. RESULTS Microarray analysis revealed significant differences in expression levels in senescent cells vs non-senescent cells: 292 genes were upregulated, and 321 downregulated. Genes with established relationships to senescence were found to be significantly upregulated in senescent cells vs. non-senescent cells: p38 (MPAK14), RB-Associated KRAB zinc finger, Discoidin, CUB and LCCL domain, growth arrest and DNA-damage inducible beta, p28ING5, sphingosine-1-phosphate receptor 2 and somatostatin receptor 3; cyclin-dependent kinase 8 showed significant downregulation in senescent cells. Nitric oxidase synthase 1, and heat shock 70 kDa protein 6, both of which were significantly down-regulated in senescent cells, also showed significant changes. Additional genes related to cytokines, cell proliferation, and other processes were also identified. CONCLUSIONS Our LCM-microarray analyses identified a set of genes associated with senescence which were significantly upregulated in senescent vs non-senescent cells in the human annulus. These genes include p38 MAP kinase, discoidin, inhibitor of growth family member 5, and growth arrest and DNA-damage-inducible beta. Other genes, including genes associated with cell proliferation, extracellular matrix formation, cell signaling and other cell functions also showed significant modulation in senescent vs non-senescent cells. The aging/degenerating disc undergoes a well-recognized loss of cells; understanding senescent cells is important since their presence further reduces the disc's ability to generate new cells to replace those lost to necrosis or apoptosis.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, NC, USA.
| | | | | | | | | |
Collapse
|
79
|
Hayashi T, Iguchi A. Possibility of the regression of atherosclerosis through the prevention of endothelial senescence by the regulation of nitric oxide and free radical scavengers. Geriatr Gerontol Int 2010; 10:115-30. [PMID: 20100288 DOI: 10.1111/j.1447-0594.2009.00581.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the elderly, atherosclerotic diseases such as stroke and myocardial infarction occupy a major part of their causes of death and care. The elderly always have atherosclerosis in their aorta and other arteries and are exposed to risk of attacks. It is the elderly who should receive its safe, harmless and advanced treatment. Advanced stage of atherosclerosis in the elderly is progressed by complicated risk factors such as dyslipidemia and diabetes mellitus and specific risk factors for the elderly, aging (and menopause). Treatment of atherosclerotic disease may need special ones targeted for the elderly. Recent studies reported that frequencies of dyslipidemia were not decreased in the older oldest. In the elderly, impaired glucose tolerance occurs and it progresses atherosclerosis. Endothelial dysfunction like impairment of nitric oxide (NO) bioavailability also progresses atherosclerosis. Although we tried to regress the high cholesterol diet-induced atherosclerosis in rabbit aorta with a normal diet with or without statin, regression could not be achieved. NO targeting gene therapy (adenovirus endothelial nitric oxide synthase [eNOS] gene vector) regressed 20% of atherosclerotic lesions through reduction of lipid contents, however, a more integrated strategy is important for complete regression. We paid attention to NO bioavailability and developed two ways of increasing it in atherosclerosis: citrulline therapy and arginase II inhibition by estrogen. Further, we found a close relation between atherosclerosis and endothelial senescence and that NO can prevent it, especially in a diabetic model. Taken together, regression of atherosclerosis can be achieved by not only regulation of various risk factors but regulation of the cross-talk of NO and free radicals.
Collapse
Affiliation(s)
- Toshio Hayashi
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | |
Collapse
|
80
|
Nitric oxide synthase inhibition aggravates the adverse renal effects of high but not low intraabdominal pressure. Surg Endosc 2009; 24:826-33. [DOI: 10.1007/s00464-009-0672-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 07/20/2009] [Accepted: 07/27/2009] [Indexed: 12/28/2022]
|
81
|
Illi B, Colussi C, Grasselli A, Farsetti A, Capogrossi MC, Gaetano C. NO sparks off chromatin: tales of a multifaceted epigenetic regulator. Pharmacol Ther 2009; 123:344-52. [PMID: 19464317 DOI: 10.1016/j.pharmthera.2009.05.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 05/11/2009] [Indexed: 10/20/2022]
Abstract
The discovery of nitric oxide (NO) revealed its ambiguous nature, which is related to its pleiotropic activities that control the homeostasis of every organism from bacteria to mammals in several physiological and pathological situations. The wide range of action of NO basically depends on two features: 1) the variety of chemical reactions depending on NO, and 2) the differential cellular responses elicited by distinct NO concentrations. Despite the increasing body of knowledge regarding its chemistry, biology and NO-dependent signaling pathways, little information is available on the nuclear actions of NO in terms of gene expression regulation. Indeed, studies of a putative role for this diatomic compound in regulating chromatin remodeling are still in their infancy. Only recently has the role of NO in epigenetics emerged, and some of its putative epigenetic properties are still only hypothetical. In the present review, we discuss the current evidence for NO-related mechanisms of epigenetic gene expression regulation. We link some of the well known NO chemical reactions and metabolic processes (e.g., S-nitrosylation of thiols, tyrosine nitration, cGMP production) to chromatin modification and address the most recent, striking hypothesis about NO and the control of chromosomes structure.
Collapse
Affiliation(s)
- Barbara Illi
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | | | | | | |
Collapse
|