301
|
Battson ML, Lee DM, Gentile CL. Endoplasmic reticulum stress and the development of endothelial dysfunction. Am J Physiol Heart Circ Physiol 2017; 312:H355-H367. [DOI: 10.1152/ajpheart.00437.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022]
Abstract
The vascular endothelium plays a critical role in cardiovascular homeostasis, and thus identifying the underlying causes of endothelial dysfunction has important clinical implications. In this regard, the endoplasmic reticulum (ER) has recently emerged as an important regulator of metabolic processes. Dysfunction within the ER, broadly termed ER stress, evokes the unfolded protein response (UPR), an adaptive pathway that aims to restore ER homeostasis. Although the UPR is the first line of defense against ER stress, chronic activation of the UPR leads to cell dysfunction and death and has recently been implicated in the pathogenesis of endothelial dysfunction. Numerous risk factors for endothelial dysfunction can induce ER stress, which may in turn disrupt endothelial function via direct effects on endothelium-derived vasoactive substances or by activating other pathogenic cellular networks such as inflammation and oxidative stress. This review summarizes the available data linking ER stress to endothelial dysfunction.
Collapse
Affiliation(s)
- M. L. Battson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| | - D. M. Lee
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| | - C. L. Gentile
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
302
|
Yin Y, Zhou Z, Liu W, Chang Q, Sun G, Dai Y. Vascular endothelial cells senescence is associated with NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation via reactive oxygen species (ROS)/thioredoxin-interacting protein (TXNIP) pathway. Int J Biochem Cell Biol 2017; 84:22-34. [PMID: 28064010 DOI: 10.1016/j.biocel.2017.01.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/12/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022]
Abstract
Endothelial dysfunction caused by endothelial cells senescence and chronic inflammation is tightly linked to the development of cardiovascular diseases. NLRP3 (NOD-like receptor family pyrin domain-containing3) inflammasome plays a central role in inflammatory response that is associated with diverse inflammatory diseases. This study explores the effects and possible mechanisms of NLRP3 inflammasome in endothelial cells senescence. Results show an increment of pro-inflammatory cytokine interleukin (IL) -1β secretion and caspase-1 activation during the senescence of endothelial cells induced by bleomycin. Moreover, secreted IL-1β promoted endothelial cells senescence through up-regulation of p53/p21 protein expression. NLRP3 inflammasome was found to mediate IL-1β secretion through the production of ROS (reactive oxygen species) during the senescence of endothelial cells. Furthermore, the association of TXNIP (thioredoxin-interacting protein) with NLRP3 induced by ROS promoted NLRP3 inflammasome activation in senescent endothelial cells. In addition, the expressions of NLRP3 inflammasome related genes, ASC (apoptosis associated speck-like protein containing a CARD), TXNIP, cleaved caspase-1 and IL-1β, were also increased in vitro and in vivo studies. These findings indicate that endothelial senescence could be mediated through ROS and NLRP3 inflammasome signaling pathways, suggesting a potential target for the prevention of endothelial senescence-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yanlin Yin
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China
| | - Zhihui Zhou
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China
| | - Weiwei Liu
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China
| | - Qun Chang
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China
| | - Guanqun Sun
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China
| | - Yalei Dai
- Department of Cardiology, Shanghai East Hospital, and Immunology Department, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
303
|
Santos-Parker JR, Strahler TR, Bassett CJ, Bispham NZ, Chonchol MB, Seals DR. Curcumin supplementation improves vascular endothelial function in healthy middle-aged and older adults by increasing nitric oxide bioavailability and reducing oxidative stress. Aging (Albany NY) 2017; 9:187-208. [PMID: 28070018 PMCID: PMC5310664 DOI: 10.18632/aging.101149] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/20/2016] [Indexed: 05/15/2023]
Abstract
We hypothesized that curcumin would improve resistance and conduit artery endothelial function and large elastic artery stiffness in healthy middle-aged and older adults. Thirty-nine healthy men and postmenopausal women (45-74 yrs) were randomized to 12 weeks of curcumin (2000 mg/day Longvida®; n=20) or placebo (n=19) supplementation. Forearm blood flow response to acetylcholine infusions (FBFACh; resistance artery endothelial function) increased 37% following curcumin supplementation (107±13 vs. 84±11 AUC at baseline, P=0.03), but not placebo (P=0.2). Curcumin treatment augmented the acute reduction in FBFACh induced by the nitric oxide synthase inhibitor NG monomethyl-L-arginine (L-NMMA; P=0.03), and reduced the acute increase in FBFACh to the antioxidant vitamin C (P=0.02), whereas placebo had no effect (both P>0.6). Similarly, brachial artery flow-mediated dilation (conduit artery endothelial function) increased 36% in the curcumin group (5.7±0.4 vs. 4.4±0.4% at baseline, P=0.001), with no change in placebo (P=0.1). Neither curcumin nor placebo influenced large elastic artery stiffness (aortic pulse wave velocity or carotid artery compliance) or circulating biomarkers of oxidative stress and inflammation (all P>0.1). In healthy middle-aged and older adults, 12 weeks of curcumin supplementation improves resistance artery endothelial function by increasing vascular nitric oxide bioavailability and reducing oxidative stress, while also improving conduit artery endothelial function.
Collapse
Affiliation(s)
| | - Talia R. Strahler
- Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Candace J. Bassett
- Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Nina Z. Bispham
- Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Michel B. Chonchol
- Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA
| | - Douglas R. Seals
- Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
304
|
Khan SY, Awad EM, Oszwald A, Mayr M, Yin X, Waltenberger B, Stuppner H, Lipovac M, Uhrin P, Breuss JM. Premature senescence of endothelial cells upon chronic exposure to TNFα can be prevented by N-acetyl cysteine and plumericin. Sci Rep 2017; 7:39501. [PMID: 28045034 PMCID: PMC5206708 DOI: 10.1038/srep39501] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is characterized by a permanent cell-cycle arrest and a pro-inflammatory secretory phenotype, and can be induced by a variety of stimuli, including ionizing radiation, oxidative stress, and inflammation. In endothelial cells, this phenomenon might contribute to vascular disease. Plasma levels of the inflammatory cytokine tumor necrosis factor alpha (TNFα) are increased in age-related and chronic conditions such as atherosclerosis, rheumatoid arthritis, psoriasis, and Crohn's disease. Although TNFα is a known activator of the central inflammatory mediator NF-κB, and can induce the intracellular generation of reactive oxygen species (ROS), the question whether TNFα can induce senescence has not been answered conclusively. Here, we investigated the effect of prolonged TNFα exposure on the fate of endothelial cells and found that such treatment induced premature senescence. Induction of endothelial senescence was prevented by the anti-oxidant N-acetyl cysteine, as well as by plumericin and PHA-408, inhibitors of the NF-κB pathway. Our results indicated that prolonged TNFα exposure could have detrimental consequences to endothelial cells by causing senescence and, therefore, chronically increased TNFα levels might possibly contribute to the pathology of chronic inflammatory diseases by driving premature endothelial senescence.
Collapse
Affiliation(s)
- Shafaat Y. Khan
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Department of Zoology, University of Sargodha, 40100 Sargodha Pakistan
| | - Ezzat M. Awad
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Andre Oszwald
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London SE5 9NU, UK
| | - Xiaoke Yin
- King’s British Heart Foundation Centre, King’s College London, London SE5 9NU, UK
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Lipovac
- Karl Landsteiner Institute for Cell-based Therapy in Gynecology, 2100 Korneuburg, Austria
| | - Pavel Uhrin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes M. Breuss
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
305
|
Altara R, Giordano M, Nordén ES, Cataliotti A, Kurdi M, Bajestani SN, Booz GW. Targeting Obesity and Diabetes to Treat Heart Failure with Preserved Ejection Fraction. Front Endocrinol (Lausanne) 2017; 8:160. [PMID: 28769873 PMCID: PMC5512012 DOI: 10.3389/fendo.2017.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major unmet medical need that is characterized by the presence of multiple cardiovascular and non-cardiovascular comorbidities. Foremost among these comorbidities are obesity and diabetes, which are not only risk factors for the development of HFpEF, but worsen symptoms and outcome. Coronary microvascular inflammation with endothelial dysfunction is a common denominator among HFpEF, obesity, and diabetes that likely explains at least in part the etiology of HFpEF and its synergistic relationship with obesity and diabetes. Thus, pharmacological strategies to supplement nitric oxide and subsequent cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling may have therapeutic promise. Other potential approaches include exercise and lifestyle modifications, as well as targeting endothelial cell mineralocorticoid receptors, non-coding RNAs, sodium glucose transporter 2 inhibitors, and enhancers of natriuretic peptide protective NO-independent cGMP-initiated and alternative signaling, such as LCZ696 and phosphodiesterase-9 inhibitors. Additionally, understanding the role of adipokines in HFpEF may lead to new treatments. Identifying novel drug targets based on the shared underlying microvascular disease process may improve the quality of life and lifespan of those afflicted with both HFpEF and obesity or diabetes, or even prevent its occurrence.
Collapse
Affiliation(s)
- Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Raffaele Altara,
| | - Mauro Giordano
- Department of Medical, Surgical, Neurological, Metabolic and Geriatrics Sciences, University of Campania “L. Vanvitelli”, Caserta, Italy
| | - Einar S. Nordén
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
| | - Mazen Kurdi
- Faculty of Sciences, Department of Chemistry and Biochemistry, Lebanese University, Hadath, Lebanon
| | - Saeed N. Bajestani
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Ophthalmology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
306
|
Hsieh PN, Sweet DR, Fan L, Jain MK. Aging and the Krüppel-like factors. TRENDS IN CELL & MOLECULAR BIOLOGY 2017; 12:1-15. [PMID: 29416266 PMCID: PMC5798252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The mammalian Krüppel-like factors (KLFs) are a family of zinc-finger containing transcription factors with diverse patterns of expression and a wide array of cellular functions. While their roles in mammalian physiology are well known, there is a growing appreciation for their roles in modulating the fundamental progression of aging. Here we review the current knowledge of Krüppel-like factors with a focus on their roles in processes regulating aging and age-associated diseases.
Collapse
Affiliation(s)
- Paishiun N. Hsieh
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - David R. Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Liyan Fan
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Mukesh K. Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
307
|
Alund AW, Mercer KE, Pulliam CF, Suva LJ, Chen JR, Badger TM, Ronis MJJ. Partial Protection by Dietary Antioxidants Against Ethanol-Induced Osteopenia and Changes in Bone Morphology in Female Mice. Alcohol Clin Exp Res 2016; 41:46-56. [PMID: 27987315 DOI: 10.1111/acer.13284] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic alcohol consumption leads to increased fracture risk and an elevated risk of osteoporosis by decreasing bone accrual through increasing osteoclast activity and decreasing osteoblast activity. We have shown that this mechanism involves the generation of reactive oxygen species (ROS) produced by NADPH oxidases. It was hypothesized that different dietary antioxidants, N-acetyl cysteine (NAC; 1.2 mg/kg/d), and α-tocopherol (Vit.E; 60 mg/kg/d) would be able to attenuate the NADPH oxidase-mediated ROS effects on bone due to chronic alcohol intake. METHODS To study the effects of these antioxidants, female mice received a Lieber-DeCarli liquid diet containing ethanol (EtOH) with or without additional antioxidant for 8 weeks. RESULTS Tibias displayed decreased cortical bone mineral density in both the EtOH and EtOH + antioxidant groups compared to pair-fed (PF) and PF + antioxidant groups (p < 0.05). However, there was significant protection from trabecular bone loss in mice fed either antioxidant (p < 0.05). Microcomputed tomography analysis demonstrated a significant decrease in bone volume (bone volume/tissue volume) and trabecular number (p < 0.05), along with a significant increase in trabecular separation in the EtOH compared to PF (p < 0.05). In contrast, the EtOH + NAC and EtOH + Vit.E did not statistically differ from their respective PF controls. Ex vivo histologic sections of tibias were stained for nitrotyrosine, an indicator of intracellular damage by ROS, and tibias from mice fed EtOH exhibited significantly more staining than PF controls. EtOH treatment significantly increased the number of marrow adipocytes per mm as well as mRNA expression of aP2, an adipocyte marker in bone. Only NAC was able to reduce the number of marrow adipocytes to PF levels. EtOH-fed mice exhibited reduced bone length (p < 0.05) and had a reduced number of proliferating chondrocytes within the growth plate. NAC and Vit.E prevented this (p < 0.05). CONCLUSIONS These data show that alcohol's pathological effects on bone extend beyond decreasing bone mass and suggest a partial protective effect of the dietary antioxidants NAC and Vit.E at these doses with regard to alcohol effects on bone turnover and bone morphology.
Collapse
Affiliation(s)
- Alexander W Alund
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Interdisciplinary Biomedical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kelly E Mercer
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Casey F Pulliam
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| | - Larry J Suva
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jin-Ran Chen
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Martin J J Ronis
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| |
Collapse
|
308
|
Martens CR, Seals DR. Practical alternatives to chronic caloric restriction for optimizing vascular function with ageing. J Physiol 2016; 594:7177-7195. [PMID: 27641062 DOI: 10.1113/jp272348] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/09/2016] [Indexed: 12/26/2022] Open
Abstract
Calorie restriction (CR) in the absence of malnutrition exerts a multitude of physiological benefits with ageing in model organisms and in humans including improvements in vascular function. Despite the well-known benefits of chronic CR, long-term energy restriction is not likely to be a feasible healthy lifestyle strategy in humans due to poor sustained adherence, and presents additional concerns if applied to normal weight older adults. This review summarizes what is known about the effects of CR on vascular function with ageing including the underlying molecular 'energy- and nutrient-sensing' mechanisms, and discusses the limited but encouraging evidence for alternative pharmacological and lifestyle interventions that may improve vascular function with ageing by mimicking the beneficial effects of long-term CR.
Collapse
Affiliation(s)
- Christopher R Martens
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
309
|
Bukiya AN, Seleverstov O, Bisen S, Dopico AM. Age-Dependent Susceptibility to Alcohol-Induced Cerebral Artery Constriction. JOURNAL OF DRUG AND ALCOHOL RESEARCH 2016; 5:236002. [PMID: 29391966 PMCID: PMC5790172 DOI: 10.4303/jdar/236002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Age has been recognized as an important contributor into susceptibility to alcohol-driven pathology. PURPOSE We aimed at determining whether alcohol-induced constriction of cerebral arteries was age-dependent. STUDY DESIGN We used rat middle cerebral artery (MCA) in vitro diameter monitoring, patch-clamping and fluorescence labeling of myocytes to study an age-dependent increase in the susceptibility to alcohol in 3 (50 g), 8 (250 g), and 15 (440 g) weeks-old rats. RESULTS An age-dependent increase in alcohol-induced constriction of MCA could be observed in absence of endothelium, which is paralleled by an age-dependent increase in both protein level of the calcium-/voltage-gated potassium channel of large conductance (BK) accessory β1 subunit and basal BK channel activity. Ethanol-induced BK channel inhibition is increased with age. CONCLUSIONS We demonstrate an increased susceptibility of MCA to ethanol-induced constriction in a period equivalent to adolescence and early adulthood when compared to pre-adolescence. Our work suggests that BK β1 constitutes a significant contributor to age-dependent changes in the susceptibility of cerebral arteries to ethanol.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Olga Seleverstov
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Shivantika Bisen
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
310
|
Basello K, Pacifici F, Capuani B, Pastore D, Lombardo MF, Ferrelli F, Coppola A, Donadel G, Arriga R, Sconocchia G, Bellia A, Rogliani P, Federici M, Sbraccia P, Lauro D, Della-Morte D. Serum- and Glucocorticoid-Inducible Kinase 1 Delay the Onset of Endothelial Senescence by Directly Interacting with Human Telomerase Reverse Transcriptase. Rejuvenation Res 2016; 19:79-89. [PMID: 26230157 DOI: 10.1089/rej.2015.1726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Endothelial senescence is characteristic of vascular aging. Serum- and glucocorticoid-inducible kinase (SGK)1 belongs to a family of serine/threonine kinases regulated by various external stimuli. SGK1 has been shown to be protective against reactive oxygen species (ROS) production and to be involved in processes regulating aging. However, data on the direct relationship between SGK1 and senescence are sparse. In the present study, we sought to investigate the role of SGK1 in cellular aging by using human umbilical vein endothelial cells (HUVECs) infected with different constructs. Senescence was measured at different cellular stages by senescence-associated β-galactosidase (SA-β-gal) activity, human telomerase reverse transcriptase (hTERT) activity, p21 protein levels, and ROS production. HUVECs over-expressing full-length SGK1 (wild-type SGK1 [SGK1WT]) showed a decrease in SA-β-gal and p21 expression and a corresponding increase in hTERT activity in the early stages of aging. Moreover, SGK1WT presented lower levels of ROS production. A direct interaction between SGK1WT and hTERT was also shown by co-immunoprecipitation. The SGK1Δ60 isoform, lacking the amino-terminal 60 amino acids, did not show interaction with hTERT, suggesting a pivotal role of this protein site for the SGK1 anti-aging function. The results from this study may be of particular importance, because SGK1WT over-expression by activating telomerase and reducing ROS levels may delay the processes of endothelial senescence.
Collapse
Affiliation(s)
- Katia Basello
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Francesca Pacifici
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Barbara Capuani
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Donatella Pastore
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Marco F Lombardo
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Francesca Ferrelli
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Andrea Coppola
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Giulia Donadel
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Roberto Arriga
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Giuseppe Sconocchia
- 2 Institute of Translational Pharmacology , National Research Council, Rome, Italy
| | - Alfonso Bellia
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Paola Rogliani
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Massimo Federici
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Paolo Sbraccia
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - Davide Lauro
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy
| | - David Della-Morte
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Italy .,3 IRCCS San Raffaele Pisana , Rome, Italy
| |
Collapse
|
311
|
Abbas M, Jesel L, Auger C, Amoura L, Messas N, Manin G, Rumig C, León-González AJ, Ribeiro TP, Silva GC, Abou-Merhi R, Hamade E, Hecker M, Georg Y, Chakfe N, Ohlmann P, Schini-Kerth VB, Toti F, Morel O. Endothelial Microparticles From Acute Coronary Syndrome Patients Induce Premature Coronary Artery Endothelial Cell Aging and Thrombogenicity: Role of the Ang II/AT1 Receptor/NADPH Oxidase-Mediated Activation of MAPKs and PI3-Kinase Pathways. Circulation 2016; 135:280-296. [PMID: 27821539 DOI: 10.1161/circulationaha.116.017513] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/19/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Microparticles (MPs) have emerged as a surrogate marker of endothelial dysfunction and cardiovascular risk. This study examined the potential of MPs from senescent endothelial cells (ECs) or from patients with acute coronary syndrome (ACS) to promote premature EC aging and thrombogenicity. METHODS Primary porcine coronary ECs were isolated from the left circumflex coronary artery. MPs were prepared from ECs and venous blood from patients with ACS (n=30) and from healthy volunteers (n=4) by sequential centrifugation. The level of endothelial senescence was assessed as senescence-associated β-galactosidase activity using flow cytometry, oxidative stress using the redox-sensitive probe dihydroethidium, tissue factor activity using an enzymatic Tenase assay, the level of target protein expression by Western blot analysis, platelet aggregation using an aggregometer, and shear stress using a cone-and-plate viscometer. RESULTS Senescence, as assessed by senescence-associated β-galactosidase activity, was induced by the passaging of porcine coronary artery ECs from passage P1 to P4, and was associated with a progressive shedding of procoagulant MPs. Exposure of P1 ECs to MPs shed from senescent P3 cells or circulating MPs from ACS patients induced increased senescence-associated β-galactosidase activity, oxidative stress, early phosphorylation of mitogen-activated protein kinases and Akt, and upregulation of p53, p21, and p16. Ex vivo, the prosenescent effect of circulating MPs from ACS patients was evidenced only under conditions of low shear stress. Depletion of endothelial-derived MPs from ACS patients reduced the induction of senescence. Prosenescent MPs promoted EC thrombogenicity through tissue factor upregulation, shedding of procoagulant MPs, endothelial nitric oxide synthase downregulation, and reduced nitric oxide-mediated inhibition of platelet aggregation. These MPs exhibited angiotensin-converting enzyme activity and upregulated AT1 receptors and angiotensin-converting enzyme in P1 ECs. Losartan, an AT1 receptor antagonist, and inhibitors of either mitogen-activated protein kinases or phosphoinositide 3-kinase prevented the MP-induced endothelial senescence. CONCLUSIONS These findings indicate that endothelial-derived MPs from ACS patients induce premature endothelial senescence under atheroprone low shear stress and thrombogenicity through angiotensin II-induced redox-sensitive activation of mitogen-activated protein kinases and phosphoinositide 3-kinase/Akt. They further suggest that targeting endothelial-derived MP shedding and their bioactivity may be a promising therapeutic strategy to limit the development of an endothelial dysfunction post-ACS.
Collapse
Affiliation(s)
- Malak Abbas
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Laurence Jesel
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Cyril Auger
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Lamia Amoura
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Nathan Messas
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Guillaume Manin
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Cordula Rumig
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Antonio J León-González
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Thais P Ribeiro
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Grazielle C Silva
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Raghida Abou-Merhi
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Eva Hamade
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Markus Hecker
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Yannick Georg
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Nabil Chakfe
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Patrick Ohlmann
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Valérie B Schini-Kerth
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Florence Toti
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.)
| | - Olivier Morel
- From UMR CNRS 7213 Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France (M.A., L.J., C.A., L.A., A.J.L.-G., T.P.R., G.C.S., V.B.S.-K., F.T., O.M.); EA7293 Stress Vasculaire et Tissulaire en Transplantation, Faculté de Pharmacie, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France (M.A., L.J., L.A.); Faculté des Sciences I. Laboratoire Génomique et Santé, Plateforme de Recherche en Sciences et Technologies, Université Libanaise, Hadath, Lebanon )M.A., R.A.-M., E.H.); Pôle d'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Fédération de Médecine Translationnelle de Strasbourg, France (N.M., G.M., Y.G., N.C., P.O., O.M.); and Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Germany (C.R., M.H.).
| |
Collapse
|
312
|
Kuang DB, Zhou JP, Yu LY, Zeng WJ, Xiao J, Zhu GZ, Zhang ZL, Chen XP. DDAH1-V3 transcript might act as miR-21 sponge to maintain balance of DDAH1-V1 in cultured HUVECs. Nitric Oxide 2016; 60:59-68. [DOI: 10.1016/j.niox.2016.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/11/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
|
313
|
Sirt1 expression is associated with CD31 expression in blood cells from patients with chronic obstructive pulmonary disease. Respir Res 2016; 17:139. [PMID: 27784320 PMCID: PMC5081972 DOI: 10.1186/s12931-016-0452-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Background Cigarette smoke induced oxidative stress has been shown to reduce silent information regulator 1 (Sirt1) levels in lung tissue from smokers and patients with COPD patients. Sirt1 is known to inhibit endothelial senescence and may play a protective role in vascular cells. Endothelial progenitor cells (EPCs) are mobilized into circulation under various pathophysiological conditions, and are thought to play an important role in tissue repair in chronic obstructive lung disease (COPD). Therefore, Sirt1 and EPC-associated mRNAs were measured in blood samples from patients with COPD and from cultured CD34+ progenitor cells to examine whether these genes are associated with COPD development. Methods This study included 358 patients with a smoking history of more than 10 pack-years. RNA was extracted from blood samples and from CD34+ progenitor cells treated with cigarette smoke extract (CSE), followed by assessment of CD31, CD34, Sirt1 mRNA, miR-34a, and miR-126-3p expression by real-time RT-PCR. Results The expression of CD31, CD34, Sirt1 mRNAs, and miR-126-3p decreased and that of miR-34a increased in moderate COPD compared with that in control smokers. However, no significant differences in these genes were observed in blood cells from patients with severe COPD compared with those in control smokers. CSE significantly decreased Sirt1 and increased miR-34a expression in cultured progenitor cells. Conclusion Sirt1 expression in blood cells from patients with COPD could be a biomarker for disease stability in patients with moderate COPD. MiR-34a may participate in apoptosis and/or senescence of EPCs in smokers. Decreased expression of CD31, CD34, and miR-126-3p potentially represents decreased numbers of EPCs in blood cell from patients with COPD.
Collapse
|
314
|
Priebe MG, McMonagle JR. Effects of Ready-to-Eat-Cereals on Key Nutritional and Health Outcomes: A Systematic Review. PLoS One 2016; 11:e0164931. [PMID: 27749919 PMCID: PMC5066953 DOI: 10.1371/journal.pone.0164931] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/04/2016] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND In many countries breakfast cereals are an important component of breakfast. This systematic review assesses the contribution of consumption of ready-to eat cereal (RTEC) to the recommended nutrient intake. Furthermore, the effects of RTEC consumption on key health parameters are investigated as well as health promoting properties of RTEC. METHOD The Cochrane Central Register of Controlled Trials, MEDLINE, EMBASE and CINAHL have been searched up till 16th of June 2015. Randomized controlled trials were excluded if RTEC were used during hypocaloric diets, if RTEC were eaten at other times than breakfast and if breakfasts included other products than RTEC, milk and fruit. Observational studies were excluded when "breakfast cereals" were not defined or their definition included cooked cereals. From cross-sectional studies only data concerning energy and nutrient intake as well as micronutrient status were used. RESULTS From 4727 identified citations 64 publications met the inclusion criteria of which 32 were cross-sectional studies, eight prospective studies and 24 randomized controlled trials. Consumption of RTEC is associated with a healthier dietary pattern, concerning intake of carbohydrates, dietary fiber, fat and micronutrients, however total sugar intake is higher. Persons consuming RTEC frequently (≥ 5 times/week) have a lower risk of inadequate micronutrient intake especially for vitamin A, calcium, folate, vitamin B 6, magnesium and zinc. Evidence from prospective studies suggests that whole grain RTEC may have beneficial effects on hypertension and type 2 diabetes. Consumption of RTEC with soluble fiber helps to reduce LDL cholesterol in hypercholesterolemic men and RTEC fortified with folate can reduce plasma homocysteine. DISCUSSION One of the review's strengths is its thorough ex/inclusion of studies. Limitations are that results of observational studies were based on self-reported data and that many studies were funded by food-industry. CONCLUSION Consumption of RTEC, especially of fiber-rich or whole grain RTEC, is implicated with several beneficial nutritional and health outcomes. The effect on body weight, intestinal health and cognitive function needs further evaluation. Of concern is the higher total sugar intake associated with frequent RTEC consumption.
Collapse
Affiliation(s)
- Marion G. Priebe
- University Medical Center Groningen, University of Groningen, Center for Medical Biomics, Groningen, The Netherlands
- Nutrition Reviewed, Murnau, Germany
| | | |
Collapse
|
315
|
Growth and maturation of heart valves leads to changes in endothelial cell distribution, impaired function, decreased metabolism and reduced cell proliferation. J Mol Cell Cardiol 2016; 100:72-82. [PMID: 27756541 DOI: 10.1016/j.yjmcc.2016.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 12/12/2022]
Abstract
Risk factors of heart valve disease are well defined and prolonged exposure throughout life leads to degeneration and dysfunction in up to 33% of the population. While aortic valve replacement remains the most common need for cardiovascular surgery particularly in those aged over 65, the underlying mechanisms of progressive deterioration are unknown. In other cardiovascular systems, a decline in endothelial cell integrity and function play a major role in promoting pathological changes, and while similar mechanisms have been speculated in the valves, studies to support this are lacking. The goal of this study was to examine age-related changes in valve endothelial cell (VEC) distribution, morphology, function and transcriptomes during critical stages of valve development (embryonic), growth (postnatal (PN)), maintenance (young adult) and aging (aging adult). Using a combination of in vivo mouse, and in vitro porcine assays we show that VEC function including, nitric oxide bioavailability, metabolism, endothelial-to-mesenchymal potential, membrane self-repair and proliferation decline with age. In addition, density of VEC distribution along the endothelium decreases and this is associated with changes in morphology, decreased cell-cell interactions, and increased permeability. These changes are supported by RNA-seq analysis showing that focal adhesion-, cell cycle-, and oxidative phosphorylation-associated biological processes are negatively impacted by aging. Furthermore, by performing high-throughput analysis we are able to report the differential and common transcriptomes of VECs at each time point that can provide insights into the mechanisms underlying age-related dysfunction. These studies suggest that maturation of heart valves over time is a multifactorial process and this study has identified several key parameters that may contribute to impairment of the valve to maintain critical structure-function relationships; leading to degeneration and disease.
Collapse
|
316
|
Stegemann S. Towards better understanding of patient centric drug product development in an increasingly older patient population. Int J Pharm 2016; 512:334-342. [DOI: 10.1016/j.ijpharm.2016.01.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/19/2016] [Indexed: 01/08/2023]
|
317
|
Takeda E, Suzuki Y, Sato Y. Age-associated downregulation of vasohibin-1 in vascular endothelial cells. Aging Cell 2016; 15:885-92. [PMID: 27325558 PMCID: PMC5013028 DOI: 10.1111/acel.12497] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 12/21/2022] Open
Abstract
Vasohibin-1 (VASH1) is an angiogenesis-inhibiting factor synthesized by endothelial cells (ECs) and it also functions to increase stress tolerance of ECs, which function is critical for the maintenance of vascular integrity. Here, we examined whether the expression of VASH1 would be affected by aging. We passaged human umbilical vein endothelial cells (HUVECs) and observed that VASH1 was downregulated in old HUVECs. This decrease in VASH1 expression with aging was confirmed in mice. To explore the mechanism of this downregulation, we compared the expression of microRNAs between old and young HUVECs by performing microarray analysis. Among the top 20 microRNAs that were expressed at a higher level in old HUVECs, the third highest microRNA, namely miR-22-3p, had its binding site on the 3' UTR of VASH1 mRNA. Experiments with microRNA mimic and anti-miR revealed that miR-22-3p was involved at least in part in the downregulation of VASH1 in ECs during replicative senescence. We then clarified the significance of this defective expression of VASH1 in the vasculature. When a cuff was placed around the femoral arteries of wild-type mice and VASH1-null mice, neointimal formation was augmented in the VASH1-null mice accompanied by an increase in adventitial angiogenesis, macrophage accumulation in the adventitia, and medial/neointimal proliferating cells. These results indicate that in replicative senescence, the downregulation of VASH1 expression in ECs was caused, at least in part, by the alteration of microRNA expression. Such downregulation of VASH1 might be involved in the acceleration of age-associated vascular diseases.
Collapse
Affiliation(s)
- Eichi Takeda
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| | - Yasuhiro Suzuki
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| | - Yasufumi Sato
- Department of Vascular Biology Institute of Development, Aging and Cancer Tohoku University 4‐1, Seiryo‐machi, Aoba‐ku Sendai 980‐8575 Japan
| |
Collapse
|
318
|
Kitahara S, Desaki J, Yoshii A, Matsui A, Morikawa S, Ezaki T. Electron microscopic study of capillary network remodeling in the extensor digitorum longus muscle of normal adult rat. Microscopy (Oxf) 2016; 65:508-516. [PMID: 27655937 DOI: 10.1093/jmicro/dfw040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/22/2016] [Indexed: 12/25/2022] Open
Abstract
Capillary networks demonstrate structural changes during maturation, aging, vascular disease, and cancer. Their morphological structure and function have an important influence on each other. Understanding the process of morphological vascular changes in the capillary network with advancing age may help overcome fatal vascular diseases. Aging-related structural changes of the capillary segments may accompany degeneration and regeneration of muscle fibers and serve to remodel the capillary network as a means of adapting to the changing environment. However, difficulty in obtaining human samples has hampered clarification of these microstructural changes. Herein, we examined serial ultrathin sections of capillary segments in the extensor digitorum longus muscle of normal mature (12 months old) rats in an attempt to analyze their structural changes. After bifurcation, a minimum of one capillary segment was filled with erythrocytes and was found to have fenestrations and plural endothelial disruptions, or pores, at the fenestrated portions. Some of the stagnated erythrocytes demonstrated extended protrusions, and their processes appeared to penetrate the basal lamina through the pores. These findings can also show that capillary segments are involved in partial remodeling of the capillary network. A better understanding of age-related structural changes of the capillary networks will help in fine-tuning novel vascular therapy for not only several fatal vascular diseases but also malignant tumors.
Collapse
Affiliation(s)
- Shuji Kitahara
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan.,Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, 7910295 MA, USA
| | - Junzo Desaki
- Department of Integrated Basic Medical Research, School of Medicine, 02114 Ehime University , Ehime, Japan
| | - Asuka Yoshii
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan.,Departments of Pharmacology and Experimental Therapeutics and Neurology, Boston University School of Medicine, Boston, 02118 MA, USA
| | - Aya Matsui
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, 1628666 Tokyo, Japan
| |
Collapse
|
319
|
Prattichizzo F, Giuliani A, De Nigris V, Pujadas G, Ceka A, La Sala L, Genovese S, Testa R, Procopio AD, Olivieri F, Ceriello A. Extracellular microRNAs and endothelial hyperglycaemic memory: a therapeutic opportunity? Diabetes Obes Metab 2016; 18:855-67. [PMID: 27161301 PMCID: PMC5094499 DOI: 10.1111/dom.12688] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/18/2016] [Accepted: 04/29/2016] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a major cause of cardiovascular (CV) disease. Several large clinical trials have shown that the risk for patients with diabetes of developing CV complications is only partially reduced by early, intensive glycaemic control and lifestyle interventions, and that such complications result from changes in complex, not fully explored networks that contribute to the maintenance of endothelial function. The accumulation of senescent cells and the low-grade, systemic, inflammatory status that accompanies aging (inflammaging) are involved in the development of endothelial dysfunction. Such phenomena are modulated by epigenetic mechanisms, including microRNAs (miRNAs). MiRNAs can modulate virtually all gene transcripts. They can be secreted by living cells and taken up in active form by recipient cells, providing a new communication tool between tissues and organs. MiRNA deregulation has been associated with the development and progression of a number of age-related diseases, including the enduring gene expression changes seen in patients with diabetes. We review recent evidence on miRNA changes in T2DM, focusing on the ability of diabetes-associated miRNAs to modulate endothelial function, inflammaging and cellular senescence. We also discuss the hypothesis that miRNA-containing extracellular vesicles (i.e. exosomes and microvesicles) could be harnessed to restore a 'physiological' signature capable of preventing or delaying the harmful systemic effects of T2DM.
Collapse
Affiliation(s)
- F Prattichizzo
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - A Giuliani
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - V De Nigris
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - G Pujadas
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - A Ceka
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - L La Sala
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| | - S Genovese
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| | - R Testa
- Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | - A D Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - F Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - A Ceriello
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| |
Collapse
|
320
|
Pantsulaia I, Ciszewski WM, Niewiarowska J. Senescent endothelial cells: Potential modulators of immunosenescence and ageing. Ageing Res Rev 2016; 29:13-25. [PMID: 27235855 DOI: 10.1016/j.arr.2016.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Recent studies have demonstrated that the accumulation of senescent endothelial cells may be the primary cause of cardiovascular diseases. Because of their multifunctional properties, endothelial cells actively take part in stimulating the immune system and inflammation. In addition, ageing is characterized by the progressive deterioration of immune cells and a decline in the activation of the immune response. This results in a loss of the primary function of the immune system, which is eliminating damaged/senescent cells and neutralizing potential sources of harmful inflammatory reactions. In this review, we discuss cellular senescence and the senescence-associated secretory phenotype (SASP) of endothelial cells and summarize the link between endothelial cells and immunosenescence. We describe the possibility that age-related changes in Toll-like receptors (TLRs) and microRNAs can affect the phenotypes of senescent endothelial cells and immune cells via a negative feedback loop aimed at restraining the excessive pro-inflammatory response. This review also addresses the following questions: how do senescent endothelial cells influence ageing or age-related changes in the inflammatory burden; what is the connection between ECs and immunosenescence, and what are the crucial hypothetical pathways linking endothelial cells and the immune system during ageing.
Collapse
|
321
|
Wang Y, Boerma M, Zhou D. Ionizing Radiation-Induced Endothelial Cell Senescence and Cardiovascular Diseases. Radiat Res 2016; 186:153-61. [PMID: 27387862 DOI: 10.1667/rr14445.1] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exposure to ionizing radiation induces not only apoptosis but also senescence. While the role of endothelial cell apoptosis in mediating radiation-induced acute tissue injury has been extensively studied, little is known about the role of endothelial cell senescence in the pathogenesis of radiation-induced late effects. Senescent endothelial cells exhibit decreased production of nitric oxide and expression of thrombomodulin, increased expression of adhesion molecules, elevated production of reactive oxygen species and inflammatory cytokines and an inability to proliferate and form capillary-like structures in vitro. These findings suggest that endothelial cell senescence can lead to endothelial dysfunction by dysregulation of vasodilation and hemostasis, induction of oxidative stress and inflammation and inhibition of angiogenesis, which can potentially contribute to radiation-induced late effects such as cardiovascular diseases (CVDs). In this article, we discuss the mechanisms by which radiation induces endothelial cell senescence, the roles of endothelial cell senescence in radiation-induced CVDs and potential strategies to prevent, mitigate and treat radiation-induced CVDs by targeting senescent endothelial cells.
Collapse
Affiliation(s)
- Yingying Wang
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Marjan Boerma
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| | - Daohong Zhou
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
322
|
The impact of hypertension on leukocyte telomere length: a systematic review and meta-analysis of human studies. J Hum Hypertens 2016; 31:99-105. [PMID: 27357526 DOI: 10.1038/jhh.2016.45] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/04/2016] [Accepted: 05/23/2016] [Indexed: 12/28/2022]
Abstract
Shortened leukocyte telomere length (LTL) is a novel biomarker for age and age-related diseases. Several epidemiological studies have examined the association between telomere length in surrogate tissues (for example, blood cells) and hypertension, and meanwhile the majority of studies reported an association some individual studies do not. We carried out a systematic review and meta-analysis to address the hypothesis that, in humans, telomere length is related with hypertension. Searches were conducted in Pubmed by September 2015 and reference lists of retrieved citations were hand searched. Eligible studies measured telomeres for both hypertensive and normotensive subjects. No restrictions were placed on sample size, publication type, age or gender. We calculated summary estimates using fixed and random effects meta-analysis. Publication bias and heterogeneity among studies were further tested. Meta-analyses from 3097 participants (1415 patients with hypertension and 1682 control subjects) showed a significant standardized mean difference between LTL in hypertensive patients and controls, either in the fixed (P<5 × 10-6) or the random model (P<0.005). Heterogeneity among studies was substantial (Q-statistic P-value <0.001, I2 97.73%). Sensitivity analysis indicated that no single study changed the standardized mean difference qualitatively (0.022> random model P-value >0.002). Egger's test for asymmetry of effect sizes (intercept±s.e.=-7.278±3.574; P=0.072) did not show evidence for strong study publication bias. Leukocyte telomeres may be shorter in hypertensive than in normotensive individuals. Larger studies controlling for confounder effects are needed to confirm these findings and further explore sources of heterogeneity.
Collapse
|
323
|
Kaplon RE, Hill SD, Bispham NZ, Santos-Parker JR, Nowlan MJ, Snyder LL, Chonchol M, LaRocca TJ, McQueen MB, Seals DR. Oral trehalose supplementation improves resistance artery endothelial function in healthy middle-aged and older adults. Aging (Albany NY) 2016; 8:1167-83. [PMID: 27208415 PMCID: PMC4931825 DOI: 10.18632/aging.100962] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/30/2016] [Indexed: 02/06/2023]
Abstract
We hypothesized that supplementation with trehalose, a disaccharide that reverses arterial aging in mice, would improve vascular function in middle-aged and older (MA/O) men and women. Thirty-two healthy adults aged 50-77 years consumed 100 g/day of trehalose (n=15) or maltose (n=17, isocaloric control) for 12 weeks (randomized, double-blind). In subjects with Δbody mass less than 2.3kg (5 lb.), resistance artery endothelial function, assessed by forearm blood flow to brachial artery infusion of acetylcholine (FBFACh), increased ~30% with trehalose (13.3±1.0 vs. 10.5±1.1 AUC, P=0.02), but not maltose (P=0.40). This improvement in FBFACh was abolished when endothelial nitric oxide (NO) production was inhibited. Endothelium-independent dilation, assessed by FBF to sodium nitroprusside (FBFSNP), also increased ~30% with trehalose (155±13 vs. 116±12 AUC, P=0.03) but not maltose (P=0.92). Changes in FBFACh and FBFSNP with trehalose were not significant when subjects with Δbody mass ≥ 2.3kg were included. Trehalose supplementation had no effect on conduit artery endothelial function, large elastic artery stiffness or circulating markers of oxidative stress or inflammation (all P>0.1) independent of changes in body weight. Our findings demonstrate that oral trehalose improves resistance artery (microvascular) function, a major risk factor for cardiovascular diseases, in MA/O adults, possibly through increasing NO bioavailability and smooth muscle sensitivity to NO.
Collapse
Affiliation(s)
- Rachelle E. Kaplon
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Sierra D. Hill
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Nina Z. Bispham
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | | | - Molly J. Nowlan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Laura L. Snyder
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Michel Chonchol
- Division of Renal Diseases & Hypertension, University of Colorado Denver, Aurora, CO 80045, USA
| | - Thomas J. LaRocca
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew B. McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
324
|
Bautista-Niño PK, Portilla-Fernandez E, Vaughan DE, Danser AHJ, Roks AJM. DNA Damage: A Main Determinant of Vascular Aging. Int J Mol Sci 2016; 17:E748. [PMID: 27213333 PMCID: PMC4881569 DOI: 10.3390/ijms17050748] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 01/16/2023] Open
Abstract
Vascular aging plays a central role in health problems and mortality in older people. Apart from the impact of several classical cardiovascular risk factors on the vasculature, chronological aging remains the single most important determinant of cardiovascular problems. The causative mechanisms by which chronological aging mediates its impact, independently from classical risk factors, remain to be elucidated. In recent years evidence has accumulated that unrepaired DNA damage may play an important role. Observations in animal models and in humans indicate that under conditions during which DNA damage accumulates in an accelerated rate, functional decline of the vasculature takes place in a similar but more rapid or more exaggerated way than occurs in the absence of such conditions. Also epidemiological studies suggest a relationship between DNA maintenance and age-related cardiovascular disease. Accordingly, mouse models of defective DNA repair are means to study the mechanisms involved in biological aging of the vasculature. We here review the evidence of the role of DNA damage in vascular aging, and present mechanisms by which genomic instability interferes with regulation of the vascular tone. In addition, we present potential remedies against vascular aging induced by genomic instability. Central to this review is the role of diverse types of DNA damage (telomeric, non-telomeric and mitochondrial), of cellular changes (apoptosis, senescence, autophagy), mediators of senescence and cell growth (plasminogen activator inhibitor-1 (PAI-1), cyclin-dependent kinase inhibitors, senescence-associated secretory phenotype (SASP)/senescence-messaging secretome (SMS), insulin and insulin-like growth factor 1 (IGF-1) signaling), the adenosine monophosphate-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR)-nuclear factor kappa B (NFκB) axis, reactive oxygen species (ROS) vs. endothelial nitric oxide synthase (eNOS)-cyclic guanosine monophosphate (cGMP) signaling, phosphodiesterase (PDE) 1 and 5, transcription factor NF-E2-related factor-2 (Nrf2), and diet restriction.
Collapse
Affiliation(s)
- Paula K Bautista-Niño
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Eliana Portilla-Fernandez
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Douglas E Vaughan
- Department of Medicine & Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
325
|
Nakayama H, Nishida K, Otsu K. Macromolecular Degradation Systems and Cardiovascular Aging. Circ Res 2016; 118:1577-92. [DOI: 10.1161/circresaha.115.307495] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
Aging-related cardiovascular diseases are a rapidly increasing problem worldwide. Cardiac aging demonstrates progressive decline of diastolic dysfunction of ventricle and increase in ventricular and arterial stiffness accompanied by increased fibrosis stimulated by angiotensin II and proinflammatory cytokines. Reactive oxygen species and multiple signaling pathways on cellular senescence play major roles in the process. Aging is also associated with an alteration in steady state of macromolecular dynamics including a dysfunction of protein synthesis and degradation. Currently, impaired macromolecular degradation is considered to be closely related to enhanced inflammation and be involved in the process and mechanism of cardiac aging. Herein, we review the role and mechanisms of the degradation system of intracellular macromolecules in the process and pathophysiology of cardiovascular aging.
Collapse
Affiliation(s)
- Hiroyuki Nakayama
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| | - Kazuhiko Nishida
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| | - Kinya Otsu
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| |
Collapse
|
326
|
De Falco E, Carnevale R, Pagano F, Chimenti I, Fianchini L, Bordin A, Siciliano C, Monticolo R, Equitani F, Carrizzo A, Peruzzi M, Vecchione C, Rubattu S, Sciarretta S, Frati G. Role of NOX2 in mediating doxorubicin-induced senescence in human endothelial progenitor cells. Mech Ageing Dev 2016; 159:37-43. [PMID: 27181082 DOI: 10.1016/j.mad.2016.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/09/2022]
Abstract
Senescence exerts a great impact on both biological and functional properties of circulating endothelial progenitor cells (EPCs), especially in cardiovascular diseases where the physiological process of aging is accelerated upon clinical administration of certain drugs such as doxorubicin. EPC impairment contributes to doxorubicin-induced cardiotoxicity. Doxorubicin accelerates EPC aging, although mechanisms underlying this phenomenon remain to be fully clarified. Here we investigated if Nox2 activity is able to modulate the premature senescence induced in vitro by doxorubicin in human EPCs. Results showed that in conditioned media obtained from late EPC cultures, the levels of interleukin-6, isoprostanes and nitric oxide bioavailability were increased and reduced respectively after 3h of doxorubicin treatment. These derangements returned to physiological levels when cells were co-treated with apocynin or gp91ds-tat (antioxidant and specific Nox2 inhibitors, respectively). Accordingly, Nox2 activity resulted to be activated by doxorubicin. Importantly, we found that Nox2 inhibition reduced doxorubicin-induced EPC senescence, as indicated by a lower percentage of β-gal positive EPCs. In conclusion, Nox2 activity efficiently contributes to the mechanism of oxidative stress-induced increase in premature aging conferred by doxorubicin. The importance of modulation of Nox2 in human EPCs could reveal a useful tool to restore EPC physiological function and properties.
Collapse
Affiliation(s)
- Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Francesca Pagano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Isotta Chimenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Luca Fianchini
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Antonella Bordin
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Camilla Siciliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Roberto Monticolo
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Francesco Equitani
- Transfusion Medicine and Immuno-Hematology Unit, Santa Maria Goretti Hospital, Latina, Italy.
| | - Albino Carrizzo
- Department of AngioCardioNeurology, IRCCS NeuroMed, 86077 Pozzilli, IS, Italy.
| | - Mariangela Peruzzi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy.
| | - Carmine Vecchione
- Department of AngioCardioNeurology, IRCCS NeuroMed, 86077 Pozzilli, IS, Italy.
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sant' Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; Department of AngioCardioNeurology, IRCCS NeuroMed, 86077 Pozzilli, IS, Italy.
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy; Department of AngioCardioNeurology, IRCCS NeuroMed, 86077 Pozzilli, IS, Italy.
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome polo pontino, C.so della Repubblica 79, 04100 Latina, Italy; Department of AngioCardioNeurology, IRCCS NeuroMed, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
327
|
Mahévas M, Michel M, Godeau B. How we manage immune thrombocytopenia in the elderly. Br J Haematol 2016; 173:844-56. [PMID: 27062054 DOI: 10.1111/bjh.14067] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
With prolonged life expectancy, immune thrombocytopenia (ITP) is frequent in elderly people. In this setting, ITP diagnosis is challenging because of the concern about an underlying myelodysplastic syndrome. Studies of older adults are lacking, and recommendations for treatment are based mainly on expert opinion. The therapeutic strategy differs from that for younger patients and must take into account the greater risk of bleeding and thrombosis, presence of comorbidities, possible impaired cognitive performance or poor life expectancy and concomitant medications, such as anticoagulant and antiplatelet therapy. Steroids and intravenous immunoglobulin (IVIg) therapy remain the first-line treatments in elderly patients, but prolonged treatment with steroids should be avoided and IVIg treatment may lead to renal failure. Splenectomy is less effective than in young patients and risk of thrombosis is increased. Severe co-morbidities can also contraindicate surgery. Therefore, other second-line treatments are frequently preferred. Danazol and dapsone can be an option for the less severe ITP form. Rituximab is a good option except in patients with a history of infection or with hypogammaglobulinaemia. Thrombopoietin agonists are attractive, especially for patients with severe comorbidities or with limited life expectancy but the risk of thrombosis is a concern.
Collapse
Affiliation(s)
- Matthieu Mahévas
- Service de Médecine Interne, Centre de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Bertrand Godeau
- Service de Médecine Interne, Centre de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| |
Collapse
|
328
|
Bambrick P, Tan WS, Mulcahy R, Pope GA, Cooke J. Vascular risk assessment in older adults without a history of cardiovascular disease. Exp Gerontol 2016; 79:37-45. [PMID: 26972634 DOI: 10.1016/j.exger.2016.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 01/27/2016] [Accepted: 03/01/2016] [Indexed: 12/16/2022]
Abstract
Modern cardiovascular risk prediction tools, which have their genesis in the Framingham Heart Study, have allowed more accurate risk stratification and targeting of treatments worldwide over the last seven decades. Better cardiovascular risk factor control during this time has led to a reduction in cardiovascular mortality and, at least in part, to improved life expectancy. As a result, western societies as a whole have seen a steady increase in the proportion of older persons in their populations. Unfortunately, several studies have shown that the same tools which have contributed to this increase cannot be reliably extrapolated for use in older generations. Recent work has allowed recalibration of existing models for use in older populations but these modified tools still require external validation before they can be confidently applied in clinical practice. Another complication is emerging evidence that aggressive risk factor modification in older adults, particularly more frail individuals, may actually be harmful. This review looks at currently available cardiovascular risk prediction models and the specific challenges faced with their use in older adults, followed by analysis of recent attempts at recalibration for this cohort. We discuss the issue of frailty, looking at our evolving understanding of its constituent features and various tools for its assessment. We also review work to date on the impact of frailty on cardiovascular risk modification and outline its potentially central role in determining the most sensible approach in older patients. We summarise the most promising novel markers of cardiovascular risk which may be of use in improving risk prediction in older adults in the future. These include markers of vascular compliance (such as aortic pulse wave velocity and pulse wave analysis), of endothelial function (such as flow mediated dilation, carotid intima-media thickness and coronary artery calcium scores), and also biochemical and circulating cellular markers.
Collapse
Affiliation(s)
- P Bambrick
- Department of Medicine for the Elderly, University Hospital Waterford, Republic of Ireland; Waterford Cardiovascular Research Group, Republic of Ireland.
| | - W S Tan
- Department of Medicine for the Elderly, University Hospital Waterford, Republic of Ireland
| | - R Mulcahy
- Department of Medicine for the Elderly, University Hospital Waterford, Republic of Ireland
| | - G A Pope
- Department of Medicine for the Elderly, University Hospital Waterford, Republic of Ireland
| | - J Cooke
- Department of Medicine for the Elderly, University Hospital Waterford, Republic of Ireland; Waterford Cardiovascular Research Group, Republic of Ireland
| |
Collapse
|
329
|
Boerman EM, Everhart JE, Segal SS. Advanced age decreases local calcium signaling in endothelium of mouse mesenteric arteries in vivo. Am J Physiol Heart Circ Physiol 2016; 310:H1091-6. [PMID: 26945073 DOI: 10.1152/ajpheart.00038.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/29/2016] [Indexed: 11/22/2022]
Abstract
Aging is associated with vascular dysfunction that impairs tissue perfusion, physical activity, and the quality of life. Calcium signaling in endothelial cells (ECs) is integral to vasomotor control, exemplified by localized Ca(2+) signals within EC projections through holes in the internal elastic lamina (IEL). Within these microdomains, endothelium-derived hyperpolarization is integral to smooth muscle cell (SMC) relaxation via coupling through myoendothelial gap junctions. However, the effects of aging on local EC Ca(2+) signals (and thereby signaling between ECs and SMCs) remain unclear, and these events have not been investigated in vivo. Furthermore, it is unknown whether aging affects either the number or the size of IEL holes. In the present study, we tested the hypothesis that local EC Ca(2+) signaling is impaired with advanced age along with a reduction in IEL holes. In anesthetized mice expressing a Ca(2+)-sensitive fluorescent protein (GCaMP2) selectively in ECs, our findings illustrate that for mesenteric arteries controlling splanchnic blood flow the frequency of spontaneous local Ca(2+) signals in ECs was reduced by ∼85% in old (24-26 mo) vs. young (3-6 mo) animals. At the same time, the number (and total area) of holes per square millimeter of IEL was reduced by ∼40%. We suggest that diminished signaling between ECs and SMCs contributes to dysfunction of resistance arteries with advanced age.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/aging-impairs-endothelial-ca2-signaling/.
Collapse
Affiliation(s)
- Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Jesse E Everhart
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
330
|
Allison BJ, Kaandorp JJ, Kane AD, Camm EJ, Lusby C, Cross CM, Nevin-Dolan R, Thakor AS, Derks JB, Tarry-Adkins JL, Ozanne SE, Giussani DA. Divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease. FASEB J 2016; 30:1968-75. [PMID: 26932929 PMCID: PMC5036970 DOI: 10.1096/fj.201500057] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/26/2016] [Indexed: 12/24/2022]
Abstract
Aging and developmental programming are both associated with oxidative stress and endothelial dysfunction, suggesting common mechanistic origins. However, their interrelationship has been little explored. In a rodent model of programmed cardiovascular dysfunction we determined endothelial function and vascular telomere length in young (4 mo) and aged (15 mo) adult offspring of normoxic or hypoxic pregnancy with or without maternal antioxidant treatment. We show loss of endothelial function [maximal arterial relaxation to acetylcholine (71 ± 3 vs. 55 ± 3%) and increased vascular short telomere abundance (4.2–1.3 kb) 43.0 ± 1.5 vs. 55.1 ± 3.8%) in aged vs. young offspring of normoxic pregnancy (P < 0.05). Hypoxic pregnancy in young offspring accelerated endothelial dysfunction (maximal arterial relaxation to acetylcholine: 42 ± 1%, P < 0.05) but this was dissociated from increased vascular short telomere length abundance. Maternal allopurinol rescued maximal arterial relaxation to acetylcholine in aged offspring of normoxic or hypoxic pregnancy but not in young offspring of hypoxic pregnancy. Aged offspring of hypoxic allopurinol pregnancy compared with aged offspring of untreated hypoxic pregnancy had lower levels of short telomeres (vascular short telomere length abundance 35.1 ± 2.5 vs. 48.2 ± 2.6%) and of plasma proinflammatory chemokine (24.6 ± 2.8 vs. 36.8 ± 5.5 pg/ml, P < 0.05). These data provide evidence for divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease, and aging being decelerated by antioxidants even prior to birth.—Allison, B. J., Kaandorp, J. J., Kane, A. D., Camm, E. J., Lusby, C., Cross, C. M., Nevin-Dolan, R., Thakor, A. S., Derks, J. B., Tarry-Adkins, J. L., Ozanne, S. E., Giussani, D. A. Divergence of mechanistic pathways mediating cardiovascular aging and developmental programming of cardiovascular disease.
Collapse
Affiliation(s)
- Beth J Allison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Joepe J Kaandorp
- Perinatology, University Medical Center, Utrecht, The Netherlands; and
| | - Andrew D Kane
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Emily J Camm
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ciara Lusby
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Christine M Cross
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rhianon Nevin-Dolan
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Avnesh S Thakor
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jan B Derks
- Perinatology, University Medical Center, Utrecht, The Netherlands; and
| | - Jane L Tarry-Adkins
- Metabolic Research Laboratories and Medical Reseach Council (MRC) Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Susan E Ozanne
- Metabolic Research Laboratories and Medical Reseach Council (MRC) Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Dino A Giussani
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom;
| |
Collapse
|
331
|
Hanson MA, Cooper C, Aihie Sayer A, Eendebak RJ, Clough GF, Beard JR. Developmental aspects of a life course approach to healthy ageing. J Physiol 2016; 594:2147-60. [PMID: 26518329 DOI: 10.1113/jp270579] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/30/2015] [Indexed: 12/23/2022] Open
Abstract
We examine the mechanistic basis and wider implications of adopting a developmental perspective on human ageing. Previous models of ageing have concentrated on its genetic basis, or the detrimental effects of accumulated damage, but also have raised issues about whether ageing can be viewed as adaptive itself, or is a consequence of other adaptive processes, for example if maintenance and repair processes in the period up to reproduction are traded off against later decline in function. A life course model places ageing in the context of the attainment of peak capacity for a body system, starting in early development when plasticity permits changes in structure and function induced by a range of environmental stimuli, followed by a period of decline, the rate of which depends on the peak attained as well as the later life conditions. Such path dependency in the rate of ageing may offer new insights into its modification. Focusing on musculoskeletal and cardiovascular function, we discuss this model and the possible underlying mechanisms, including endothelial function, oxidative stress, stem cells and nutritional factors such as vitamin D status. Epigenetic changes induced during developmental plasticity, and immune function may provide a common mechanistic process underlying a life course model of ageing. The life course trajectory differs in high and low resource settings. New insights into the developmental components of the life course model of ageing may lead to the design of biomarkers of later chronic disease risk and to new interventions to promote healthy ageing, with important implications for public health.
Collapse
Affiliation(s)
- M A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK.,NIHR Nutrition Biomedical Research Centre, University Hospital Southampton, UK
| | - C Cooper
- NIHR Nutrition Biomedical Research Centre, University Hospital Southampton, UK.,MRC Lifecourse Epidemiology Unit, University Hospital Southampton, UK
| | - A Aihie Sayer
- NIHR Nutrition Biomedical Research Centre, University Hospital Southampton, UK.,MRC Lifecourse Epidemiology Unit, University Hospital Southampton, UK
| | - R J Eendebak
- Andrology Research Unit, Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Old St Mary's Building, Hathersage Road, Manchester, UK
| | - G F Clough
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - J R Beard
- Department of Ageing and Lifecourse, World Health Organization, 20 Avenue Appia, 1211, Geneva 27, Switzerland
| |
Collapse
|
332
|
Verma SK, Garikipati VNS, Krishnamurthy P, Khan M, Thorne T, Qin G, Losordo DW, Kishore R. IL-10 Accelerates Re-Endothelialization and Inhibits Post-Injury Intimal Hyperplasia following Carotid Artery Denudation. PLoS One 2016; 11:e0147615. [PMID: 26808574 PMCID: PMC4725953 DOI: 10.1371/journal.pone.0147615] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022] Open
Abstract
The role of inflammation on atherosclerosis and restenosis is well established. Restenosis is thought to be a complex response to injury, which includes early thrombus formation, acute inflammation and neo-intimal growth. Inflammatory cells are likely contributors in the host response to vascular injury, via cytokines and chemokines secretion, including TNF-alpha (TNF). We have previously shown that IL-10 inhibits TNF and other inflammatory mediators produced in response to cardiovascular injuries. The specific effect of IL-10 on endothelial cell (ECs) biology is not well elucidated. Here we report that in a mouse model of carotid denudation, IL-10 knock-out mice (IL-10KO) displayed significantly delayed Re-endothelialization and enhanced neo-intimal growth compared to their WT counterparts. Exogenous recombinant IL-10 treatment dramatically blunted the neo-intimal thickening while significantly accelerating the recovery of the injured endothelium in WT mice. In vitro, IL-10 inhibited negative effects of TNF on ECs proliferation, ECs cell cycle, ECs-monocyte adhesion and ECs apoptosis. Furthermore, IL-10 treatment attenuated TNF-induced smooth muscle cells proliferation. Our data suggest that IL-10 differentially regulate endothelial and vascular smooth cells proliferation and function and thus inhibits neo-intimal hyperplasia. Thus, these results may provide insights necessary to develop new therapeutic strategies to limit vascular restenosis during percutaneous coronary intervention (PCI) in the clinics.
Collapse
Affiliation(s)
- Suresh K Verma
- Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania United States of America
| | | | - Prasanna Krishnamurthy
- Department of Cardiovascular Science, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Mohsin Khan
- Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania United States of America
| | - Tina Thorne
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Gangjian Qin
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Douglas W Losordo
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Raj Kishore
- Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania United States of America.,Department of Pharmacology, Temple University, Philadelphia, Pennsylvania United States of America
| |
Collapse
|
333
|
Gargiulo S, Gamba P, Testa G, Leonarduzzi G, Poli G. The role of oxysterols in vascular ageing. J Physiol 2016; 594:2095-113. [PMID: 26648329 DOI: 10.1113/jp271168] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/06/2015] [Indexed: 12/24/2022] Open
Abstract
The ageing endothelium progressively loses its remarkable and crucial ability to maintain homeostasis of the vasculature, as it acquires a proinflammatory phenotype. Cellular and structural changes gradually accumulate in the blood vessels, and markedly in artery walls. Most changes in aged arteries are comparable to those occurring during the atherogenic process, the latter being more marked: pro-oxidant and proinflammatory molecules, mainly deriving from or triggered by oxidized low density lipoproteins (oxLDLs), are undoubtedly a major driving force of this process. Oxysterols, quantitatively relevant components of oxLDLs, are likely candidate molecules in the pathogenesis of vascular ageing, because of their marked pro-oxidant, proinflammatory and proapoptotic properties. An increasing bulk of experimental data point to the contribution of a variety of oxysterols of pathophysiological interest, also in the age-related genesis of endothelium dysfunction, intimal thickening due to lipid accumulation, and smooth muscle cell migration and arterial stiffness due to increasing collagen deposition and calcification. This review provides an updated analysis of the molecular mechanisms whereby oxysterols accumulating in the wall of ageing blood vessels may 'activate' endothelial and monocytic cells, through expression of an inflammatory phenotype, and 'convince' smooth muscle cells to proliferate, migrate and, above all, to act as fibroblast-like cells.
Collapse
Affiliation(s)
- Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| |
Collapse
|
334
|
Gu L, Hitzel J, Moll F, Kruse C, Malik RA, Preussner J, Looso M, Leisegang MS, Steinhilber D, Brandes RP, Fork C. The Histone Demethylase PHF8 Is Essential for Endothelial Cell Migration. PLoS One 2016; 11:e0146645. [PMID: 26751588 PMCID: PMC4713448 DOI: 10.1371/journal.pone.0146645] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/20/2015] [Indexed: 01/26/2023] Open
Abstract
Epigenetic marks critically control gene expression and thus the cellular activity state. The functions of many epigenetic modifiers in the vascular system have not yet been studied. We screened for histone modifiers in endothelial cells and observed a fairly high expression of the histone plant homeodomain finger protein 8 (PHF8). Given its high expression, we hypothesize that this histone demethylase is important for endothelial cell function. Overexpression of PHF8 catalyzed the removal of methyl-groups from histone 3 lysine 9 (H3K9) and H4K20, whereas knockdown of the enzyme increased H3K9 methylation. Knockdown of PHF8 by RNAi also attenuated endothelial proliferation and survival. As a functional readout endothelial migration and tube formation was studied. PHF8 siRNA attenuated the capacity for migration and developing of capillary-like structures. Given the impact of PHF8 on cell cycle genes, endothelial E2F transcription factors were screened, which led to the identification of the gene repressor E2F4 to be controlled by PHF8. Importantly, PHF8 maintains E2F4 but not E2F1 expression in endothelial cells. Consistently, chromatin immunoprecipitation revealed that PHF8 reduces the H3K9me2 level at the E2F4 transcriptional start site, demonstrating a direct function of PHF8 in endothelial E2F4 gene regulation. Conclusion: PHF8 by controlling E2F4 expression maintains endothelial function.
Collapse
Affiliation(s)
- Lunda Gu
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
| | - Juliane Hitzel
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Franziska Moll
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Christoph Kruse
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Randa Abdel Malik
- Institute of Vascular Signalling, Centre for Molecular Medicine, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Matthias S. Leisegang
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Christian Fork
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
335
|
Modifiable Factors Influencing Telomere Length and Aging. INFLAMMATION, AGING, AND OXIDATIVE STRESS 2016. [DOI: 10.1007/978-3-319-33486-8_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
336
|
Thorin-Trescases N, Thorin E. Lifelong Cyclic Mechanical Strain Promotes Large Elastic Artery Stiffening: Increased Pulse Pressure and Old Age-Related Organ Failure. Can J Cardiol 2015; 32:624-33. [PMID: 26961664 DOI: 10.1016/j.cjca.2015.12.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/02/2015] [Accepted: 12/14/2015] [Indexed: 01/08/2023] Open
Abstract
The arterial wall is under a huge mechanical constraint imposed by the cardiac cycle that is bound to generate damage with time. Each heartbeat indeed imposes a pulsatile pressure that generates a vascular stretch. Lifetime accumulation of pulsatile stretches will eventually induce fatigue of the elastic large arterial walls, such as aortic and carotid artery walls, promoting their stiffening that will gradually perturb the normal blood flow and local pressure within the organs, and lead to organ failure. The augmented pulse pressure induced by arterial stiffening favours left ventricular hypertrophy because of the repeated extra work against stiff high-pressure arteries, and tissue damage as a result of excessive pulsatile pressure transmitted into the microcirculation, especially in low resistance/high-flow organs such as the brain and kidneys. Vascular aging is therefore characterized by the stiffening of large elastic arteries leading to a gradual increase in pulse pressure with age. In this review we focus on the effect of age-related stiffening of large elastic arteries. We report the clinical evidence linking arterial stiffness and organ failure and discuss the molecular pathways that are activated by the increase of mechanical stress in the wall. We also discuss the possible interventions that could limit arterial stiffening with age, such as regular aerobic exercise training, and some pharmacological approaches.
Collapse
Affiliation(s)
| | - Eric Thorin
- Montreal Heart Institute, Research Center, Montreal, Quebéc, Canada; Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, Quebéc, Canada.
| |
Collapse
|
337
|
Changes in Oxidative Stress and Inflammatory Biomarkers in Fragile Adults over Fifty Years of Age and in Elderly People Exclusively Fed Enteral Nutrition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:5709312. [PMID: 26697137 PMCID: PMC4678080 DOI: 10.1155/2016/5709312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/14/2015] [Accepted: 08/16/2015] [Indexed: 12/18/2022]
Abstract
We aim to evaluate whether exclusive feeding of an enteral formula enriched with n-3 long chain polyunsaturated fatty acids (n-3 LC-PUFA) affects oxidative stress and the antioxidant defence system and may improve the levels of some relevant inflammatory, and cardiovascular biomarkers in frail adults over fifty years of age and in elderly subjects. Fifty-five patients were divided into two groups and were exclusively fed a newly designed normoproteic and isocaloric enteral formula enriched with eicosapentaenoic (98 mg/d) and docosahexaenoic acids (46 mg/d) (n = 26) or a reference enteral diet (n = 29). Oxidative, inflammatory and cardiovascular risk biomarkers and red blood cell fatty acid profiles were determined at the beginning and after 90 and 180 days of feeding. The n-3 LC-PUFA percentage tended to be higher (P = 0.053) in the experimental group than in the reference group. Administration of the n-3 LC-PUFA diet did not increase oxidative stress or modify plasma antioxidant capacity but decreased antioxidant enzymatic activities. MMP-9 plasma concentration decreased with both formulae, whereas tPAI-1 tended to decrease (P = 0.116) with the administration of the experimental formula. In conclusion, administration of the new n-3 LC-PUFA-enriched product for 6 months did not negatively alter the oxidative status and improved some cardiovascular risk biomarkers.
Collapse
|
338
|
Galvan V, Hart MJ. Vascular mTOR-dependent mechanisms linking the control of aging to Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:992-1007. [PMID: 26639036 DOI: 10.1016/j.bbadis.2015.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
Aging is the strongest known risk factor for Alzheimer's disease (AD). With the discovery of the mechanistic target of rapamycin (mTOR) as a critical pathway controlling the rate of aging in mice, molecules at the interface between the regulation of aging and the mechanisms of specific age-associated diseases can be identified. We will review emerging evidence that mTOR-dependent brain vascular dysfunction, a universal feature of aging, may be one of the mechanisms linking the regulation of the rate of aging to the pathogenesis of Alzheimer's disease. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Veronica Galvan
- Department of Physiology and the Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio.
| | - Matthew J Hart
- Department of Biochemistry, University of Texas Health Science Center at San Antonio
| |
Collapse
|
339
|
DeVan AE, Johnson LC, Brooks FA, Evans TD, Justice JN, Cruickshank-Quinn C, Reisdorph N, Bryan NS, McQueen MB, Santos-Parker JR, Chonchol MB, Bassett CJ, Sindler AL, Giordano T, Seals DR. Effects of sodium nitrite supplementation on vascular function and related small metabolite signatures in middle-aged and older adults. J Appl Physiol (1985) 2015; 120:416-25. [PMID: 26607249 DOI: 10.1152/japplphysiol.00879.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/20/2015] [Indexed: 12/20/2022] Open
Abstract
Insufficient nitric oxide (NO) bioavailability plays an important role in endothelial dysfunction and arterial stiffening with aging. Supplementation with sodium nitrite, a precursor of NO, ameliorates age-related vascular endothelial dysfunction and arterial stiffness in mice, but effects on humans, including the metabolic pathways altered, are unknown. The purpose of this study was to determine the safety, feasibility, and efficacy of oral sodium nitrite supplementation for improving vascular function in middle-aged and older adults and to identify related circulating metabolites. Ten weeks of sodium nitrite (80 or 160 mg/day, capsules, TheraVasc; randomized, placebo control, double blind) increased plasma nitrite acutely (5- to 15-fold, P < 0.001 vs. placebo) and chronically (P < 0.10) and was well tolerated without symptomatic hypotension or clinically relevant elevations in blood methemoglobin. Endothelial function, measured by brachial artery flow-mediated dilation, increased 45-60% vs. baseline (P < 0.10) without changes in body mass or blood lipids. Measures of carotid artery elasticity (ultrasound and applanation tonometry) improved (decreased β-stiffness index, increased cross-sectional compliance, P < 0.05) without changes in brachial or carotid artery blood pressure. Aortic pulse wave velocity was unchanged. Nitrite-induced changes in vascular measures were significantly related to 11 plasma metabolites identified by untargeted analysis. Baseline abundance of multiple metabolites, including glycerophospholipids and fatty acyls, predicted vascular changes with nitrite. This study provides evidence that sodium nitrite supplementation is well tolerated, increases plasma nitrite concentrations, improves endothelial function, and lessens carotid artery stiffening in middle-aged and older adults, perhaps by altering multiple metabolic pathways, thereby warranting a larger clinical trial.
Collapse
Affiliation(s)
- Allison E DeVan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado;
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Forrest A Brooks
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Trent D Evans
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Nichole Reisdorph
- Integrated Department of Immunology, National Jewish Health, Denver, Colorado
| | | | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Michel B Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Candace J Bassett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Amy L Sindler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
340
|
Lamoke F, Shaw S, Yuan J, Ananth S, Duncan M, Martin P, Bartoli M. Increased Oxidative and Nitrative Stress Accelerates Aging of the Retinal Vasculature in the Diabetic Retina. PLoS One 2015; 10:e0139664. [PMID: 26466127 PMCID: PMC4605485 DOI: 10.1371/journal.pone.0139664] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 09/16/2015] [Indexed: 01/12/2023] Open
Abstract
Hyperglycemia-induced retinal oxidative and nitrative stress can accelerate vascular cell aging, which may lead to vascular dysfunction as seen in diabetes. There is no information on whether this may contribute to the progression of diabetic retinopathy (DR). In this study, we have assessed the occurrence of senescence-associated markers in retinas of streptozotocin-induced diabetic rats at 8 and 12 weeks of hyperglycemia as compared to normoglycemic aging (12 and 14 months) and adult (4.5 months) rat retinas. We have found that in the diabetic retinas there was an up-regulation of senescence-associated markers SA-β-Gal, p16INK4a and miR34a, which correlated with decreased expression of SIRT1, a target of miR34a. Expression of senescence-associated factors primarily found in retinal microvasculature of diabetic rats exceeded levels measured in adult and aging rat retinas. In aging rats, retinal expression of senescence associated-factors was mainly localized at the level of the retinal pigmented epithelium and only minimally in the retinal microvasculature. The expression of oxidative/nitrative stress markers such as 4-hydroxynonenal and nitrotyrosine was more pronounced in the retinal vasculature of diabetic rats as compared to normoglycemic aging and adult rat retinas. Treatments of STZ-rats with the anti-nitrating drug FeTPPS (10mg/Kg/day) significantly reduced the appearance of senescence markers in the retinal microvasculature. Our results demonstrate that hyperglycemia accelerates retinal microvascular cell aging whereas physiological aging affects primarily cells of the retinal pigmented epithelium. In conclusion, hyperglycemia-induced retinal vessel dysfunction and DR progression involve vascular cell senescence due to increased oxidative/nitrative stress.
Collapse
Affiliation(s)
- Folami Lamoke
- Dept. of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Sean Shaw
- Dept. of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Jianghe Yuan
- Dept. of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Sudha Ananth
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Michael Duncan
- Dept. of Medicine, Section of Gastroenterology/Hepatology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Pamela Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Manuela Bartoli
- Dept. of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
341
|
Thijssen DHJ, Carter SE, Green DJ. Arterial structure and function in vascular ageing: are you as old as your arteries? J Physiol 2015; 594:2275-84. [PMID: 26140618 DOI: 10.1113/jp270597] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/29/2015] [Indexed: 12/11/2022] Open
Abstract
Advancing age may be the most potent independent predictor of future cardiovascular events, a relationship that is not fully explained by time-related changes in traditional cardiovascular risk factors. Since some arteries exhibit differential susceptibility to atherosclerosis, generalisations regarding the impact of ageing in humans may be overly simplistic, whereas in vivo assessment of arterial function and health provide direct insight. Coronary and peripheral (conduit, resistance and skin) arteries demonstrate a gradual, age-related impairment in vascular function that is likely to be related to a reduction in endothelium-derived nitric oxide bioavailability and/or increased production of vasoconstrictors (e.g. endothelin-1). Increased exposure and impaired ability for defence mechanisms to resist oxidative stress and inflammation, but also cellular senescence processes, may contribute to age-related changes in vascular function and health. Arteries also undergo structural changes as they age. Gradual thickening of the arterial wall, changes in wall content (i.e. less elastin, advanced glycation end-products) and increase in conduit artery diameter are observed with older age and occur similarly in central and peripheral arteries. These changes in structure have important interactive effects on artery function, with increases in small and large arterial stiffness representing a characteristic change with older age. Importantly, direct measures of arterial function and structure predict future cardiovascular events, independent of age or other cardiovascular risk factors. Taken together, and given the differential susceptibility of arteries to atherosclerosis in humans, direct measurement of arterial function and health may help to distinguish between biological and chronological age-related change in arterial health in humans.
Collapse
Affiliation(s)
- Dick H J Thijssen
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, UK.,Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, The Netherlands
| | - Sophie E Carter
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, UK
| | - Daniel J Green
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, UK.,School of Sports Science, Exercise and Health, The University of Western Australia, Western Australia
| |
Collapse
|
342
|
Simultaneous Sensitive MEKC–LIF Determination of Homocysteine, Homoarginine, and Six Arginine Metabolic Derivatives in Fluids from Type 2 Diabetics with Peptic Ulcer Bleeding. Chromatographia 2015. [DOI: 10.1007/s10337-015-2919-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
343
|
Lakatta EG. So! What's aging? Is cardiovascular aging a disease? J Mol Cell Cardiol 2015; 83:1-13. [PMID: 25870157 PMCID: PMC4532266 DOI: 10.1016/j.yjmcc.2015.04.005] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 04/02/2015] [Accepted: 04/03/2015] [Indexed: 12/21/2022]
Abstract
"Inside every old person is a young person wondering what happened." So, what is aging? Aging is a manifestation of progressive, time-dependent failure of molecular mechanisms that create disorder within a system of DNA and its environment (nuclear, cytosolic, tissue, organ, organism, other organisms, society, terra firma, atmosphere, universe). Continuous signaling, transmitted with different kinetics across each of these environments, confers a "mutual enslavement" that creates ordered functions among the components within the system. Accrual of this molecular disorder over time, i.e. during aging, causes progressive changes in the structure and function of the heart and arteries that are quite similar in humans, non-human primates, rabbits and rats that compromise cardiovascular reserve function, and confer a marked risk for incident cardiovascular disease. Nearly all aspects of signaling within the DNA environment system within the heart and arteries become disordered with advancing age: Signals change, as does sensing of the signals, transmission of signals and responses to signals, impaired cell renewal, changes in the proteome due to alterations in genomic transcription, mRNA translation, and proteostasis. The density of some molecules becomes reduced, and post-translational modifications, e.g. oxidation and nitration phosphorylation, lead to altered misfolding and disordered molecular interactions. The stoichiometry and kinetics of enzymatic and those reactions which underlie crucial cardiac and vascular cell functions and robust reserve mechanisms that remove damaged organelles and proteins deteriorate. The CV cells generate an inflammatory defense in an attempt to limit the molecular disorder. The resultant proinflammatory milieu is not executed by "professional" inflammatory cells (i.e. white blood cells), however, but by activation of renin-angiotensin-aldosterone endothelin signaling cascades that leads to endothelial and vascular smooth muscle and cardiac cells' phenotype shifts, resulting in production of inflammatory cytokines. Progressive molecular disorder within the heart and arteries over time leads to an excessive allostatic load on the CV system, that results in an increase and "overshoot" in the inflammatory defense signaling. This age-associated molecular disorder-induced inflammation that accrues in the heart and arteries does not, itself, cause clinical signs or symptoms of CVD. Clinical signs and symptoms of these CVDs begin to emerge, however, when the age-associated inflammation in the heart and arteries exceeds a threshold. Thus, an emerging school of thought is that accelerated age-associated alterations within the heart and arteries, per se, ought to be considered to be a type of CVD, because the molecular disorder and the inflammatory milieu it creates within the heart and arteries with advancing age are the roots of the pathophysiology of most cardiovascular diseases, e.g. athersclerosis and hypertension. Because many effects of aging on the CV system can be delayed or attenuated by changes in lifestyle, e.g. diet and exercise, or by presently available drugs, e.g. those that suppress Ang II signaling, CV aging is a promising frontier in preventive cardiology that is not only ripe for, but also in dire need of attention! There is an urgency to incorporate the concept of cardiovascular aging as a disease into clinical medicine. But, sadly, the reality of the age-associated molecular disorder within the heart and ateries has, for the most part, been kept outside of mainstream clinical medicine. This article is part of a Special Issue entitled CV Aging.
Collapse
Affiliation(s)
- Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, Biomedical Research Center, NIH, 251 Bayview Blvd., Baltimore, MD 21224, USA.
| |
Collapse
|
344
|
Alvarado-Vásquez N. Circulating cell-free mitochondrial DNA as the probable inducer of early endothelial dysfunction in the prediabetic patient. Exp Gerontol 2015; 69:70-8. [PMID: 26026597 DOI: 10.1016/j.exger.2015.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 05/09/2015] [Accepted: 05/25/2015] [Indexed: 12/16/2022]
Abstract
Recent evidence has shown that 346million people in the world have diabetes mellitus (DM); this number will increase to 439million by 2030. In addition, current data indicate an increase in DM cases in the population between 40 and 59years of age. Diabetes is associated with the development of micro- and macro-vascular complications, derived from chronic hyperglycemia on the endothelium. Some reports demonstrate that people in a prediabetic state have a major risk of developing early endothelial dysfunction (ED). Today, it is accepted that individuals considered as prediabetic patients are in a pro-inflammatory state associated with endothelial and mitochondrial dysfunction. It is important to mention that impaired mitochondrial functionality has been linked to endothelial apoptosis and release of mitochondrial DNA (mtDNA) in patients with sepsis, cardiac disease, or atherosclerosis. This free mtDNA could promote ED, as well as other side effects on the vascular system through the activation of the toll-like receptor 9 (TLR9). TLR9 is expressed in different cell types (e.g., T or B lymphocytes, mastocytes, and epithelial and endothelial cells). It is localized intracellularly and recognizes non-methylated dinucleotides of viral, bacterial, and mitochondrial DNA. Recently, it has been reported that TLR9 is associated with the pathogenesis of lupus erythematosus, rheumatoid arthritis, and autoimmune diabetes. In this work, it is hypothesized that the increase in the levels of circulating mtDNA is the trigger of early ED in the prediabetic patient, and later on in the older patient with diabetes, through activation of the TLR9 present in the endothelium.
Collapse
Affiliation(s)
- Noé Alvarado-Vásquez
- Department of Biochemistry, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calz. de Tlalpan 4502, Col. Sección XVI, 14080 Mexico, D.F., Mexico, Mexico.
| |
Collapse
|