51
|
Lee DY, Chiu JJ. Atherosclerosis and flow: roles of epigenetic modulation in vascular endothelium. J Biomed Sci 2019; 26:56. [PMID: 31387590 PMCID: PMC6685237 DOI: 10.1186/s12929-019-0551-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background Endothelial cell (EC) dysfunctions, including turnover enrichment, gap junction disruption, inflammation, and oxidation, play vital roles in the initiation of vascular disorders and atherosclerosis. Hemodynamic forces, i.e., atherprotective pulsatile (PS) and pro-atherogenic oscillatory shear stress (OS), can activate mechanotransduction to modulate EC function and dysfunction. This review summarizes current studies aiming to elucidate the roles of epigenetic factors, i.e., histone deacetylases (HDACs), non-coding RNAs, and DNA methyltransferases (DNMTs), in mechanotransduction to modulate hemodynamics-regulated EC function and dysfunction. Main body of the abstract OS enhances the expression and nuclear accumulation of class I and class II HDACs to induce EC dysfunction, i.e., proliferation, oxidation, and inflammation, whereas PS induces phosphorylation-dependent nuclear export of class II HDACs to inhibit EC dysfunction. PS induces overexpression of the class III HDAC Sirt1 to enhance nitric oxide (NO) production and prevent EC dysfunction. In addition, hemodynamic forces modulate the expression and acetylation of transcription factors, i.e., retinoic acid receptor α and krüppel-like factor-2, to transcriptionally regulate the expression of microRNAs (miRs). OS-modulated miRs, which stimulate proliferative, pro-inflammatory, and oxidative signaling, promote EC dysfunction, whereas PS-regulated miRs, which induce anti-proliferative, anti-inflammatory, and anti-oxidative signaling, inhibit EC dysfunction. PS also modulates the expression of long non-coding RNAs to influence EC function. i.e., turnover, aligmant, and migration. On the other hand, OS enhances the expression of DNMT-1 and -3a to induce EC dysfunction, i.e., proliferation, inflammation, and NO repression. Conclusion Overall, epigenetic factors play vital roles in modulating hemodynamic-directed EC dysfunction and vascular disorders, i.e., atherosclerosis. Understanding the detailed mechanisms through which epigenetic factors regulate hemodynamics-directed EC dysfunction and vascular disorders can help us to elucidate the pathogenic mechanisms of atherosclerosis and develop potential therapeutic strategies for atherosclerosis treatment.
Collapse
Affiliation(s)
- Ding-Yu Lee
- Department of Biological Science and Technology, China University of Science and Technology, Taipei, 115, Taiwan
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 350, Taiwan. .,Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan. .,Collage of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan. .,Institute of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan. .,Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
52
|
Ji JY. Endothelial Nuclear Lamina in Mechanotransduction Under Shear Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1097:83-104. [PMID: 30315541 DOI: 10.1007/978-3-319-96445-4_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Endothelial cells that line the lumen of blood vessels are at the interface between hemodynamic forces and vascular wall biology. Endothelial cells transduce mechanical and biological signals from blood flow into intracellular signaling cascades through a process called mechanotransduction. Mechanotransduction is an important part of normal cell functions, as well as endothelial dysfunction which leads to inflammation and pathological conditions. For example, atherosclerosis preferentially develops in regions of disturbed fluid flow and low shear stress. The nuclear lamina, which sits underneath the nuclear envelope, serves to maintain the nuclear structure while acting as a scaffold for heterochromatin and many transcriptional proteins. Defects in lamina and its associated proteins cause a variety of human diseases including accelerated aging diseases such as Hutchinson-Gilford Progeria syndrome. The role of nuclear lamina in endothelial mechanotransduction, specifically how nuclear mechanics impact gene regulation under shear stress, is not fully understood. In one study, lamin A/C was silenced in bovine aortic endothelial cells to determine its role in both glucocorticoid receptor (GR) nuclear translocation and glucocorticoid response element (GRE) transcriptional activation in response to its natural ligand dexamethasone as well as fluid shear stress. Results suggest that absence of lamin A/C does not hinder passage of GR into the nucleus but nuclear lamina is important to properly regulate GRE transcription. Ongoing research continues to investigate how nuclear lamins contribute to endothelial mechanotransduction and to better understand the role of Lamin A in vascular aging and in the progression of cardiovascular diseases.
Collapse
Affiliation(s)
- Julie Y Ji
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
53
|
Dyck GJB, Raj P, Zieroth S, Dyck JRB, Ezekowitz JA. The Effects of Resveratrol in Patients with Cardiovascular Disease and Heart Failure: A Narrative Review. Int J Mol Sci 2019; 20:ijms20040904. [PMID: 30791450 PMCID: PMC6413130 DOI: 10.3390/ijms20040904] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/03/2019] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death globally and responsible for the second highest number of deaths in Canada. Medical advancements in the treatment of CVD have led to patients living longer with CVD but often progressing to another condition called heart failure (HF). As a result, HF has emerged in the last decade as a major medical concern. Fortunately, various “traditional” pharmacotherapies for HF exist and have shown success in reducing HF-associated mortality. However, to augment the treatment of patients with CVD and/or HF, alternative pharmacotherapies using nutraceuticals have also shown promise in the prevention and treatment of these two conditions. One of these natural compounds considered to potentially help treat HF and CVD and prevent their development is resveratrol. Herein, we review the clinical findings of resveratrol’s ability to be used as an effective treatment to potentially help treat HF and CVD. This will allow us to gain a more fulsome appreciation for the effects of resveratrol in the health outcomes of specific patient populations who have various disorders that constitute CVD.
Collapse
Affiliation(s)
- Garrison J B Dyck
- Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Pema Raj
- St Boniface Hospital, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.
| | - Shelley Zieroth
- St Boniface Hospital, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Justin A Ezekowitz
- Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
54
|
Naskar S, Panda AK, Kumaran V, Mehta B, Basu B. Controlled Shear Flow Directs Osteogenesis on UHMWPE-Based Hybrid Nanobiocomposites in a Custom-Designed PMMA Microfluidic Device. ACS APPLIED BIO MATERIALS 2018; 1:414-435. [DOI: 10.1021/acsabm.8b00147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sharmistha Naskar
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
- Department of Chemical Engineering, Indian Institute of Science, Bangalore 560012, India
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Asish Kumar Panda
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Viswanathan Kumaran
- Department of Chemical Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bikramjit Basu
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
55
|
Metabololipidomic profiling of functional immunoresolvent clusters and eicosanoids in mammalian tissues. Biochem Biophys Res Commun 2018. [PMID: 29524409 DOI: 10.1016/j.bbrc.2018.03.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Metabolomics enables a systems approach to interrogate the bioactive mediators, their pathways and further metabolites involved in the physiology and pathophysiology of human and animal tissues. New metabololipidomic approaches with mass spectrometry presented in this brief review can now be utilized for the identification and profiling of lipid mediator networks that control inflammation-resolution in human blood and healthy and diseased solid tissues. Coagulation of blood is a protective response that prevents excessive bleeding on injury of blood vessels. Here, we review novel approaches to understand the relationship(s) between coagulation and resolution of inflammation and infection. To determine whether coagulation is involved in host-protective actions by lipid mediators, we used a metabololipidomic-based profiling approach with human whole blood (WB) during coagulation. We identified recently temporal clusters of endogenously produced pro-thrombotic and proinflammatory lipid mediators (eicosanoids), as well as specialized proresolving mediators (SPMs) in this vital process. In addition to the classic eicosanoids (prostaglandins, thromboxanes and leukotrienes), a specific SPM cluster was identified that consists of resolvin E1 (RvE1), RvD1, RvD5, lipoxin B4, and maresin 1, each of which present at bioactive concentrations (0.1-1 nM). The removal of adenosine from coagulating blood samples significantly enhances SPM amounts and unleashes the biosynthesis of RvD3, RvD4, and RvD6 evident following rapid snap freezing with centrifugation before extraction and LC-MS-MS. The classic cyclooxygenase inhibitors, celecoxib and indomethacin, that block thromboxanes and prostanoids do not block production of the clot-driven SPM cluster. Unbiased mass cytometry analysis demonstrated that the SPM cluster produced in human blood targets leukocytes at the single-cell level, directly activating extracellular signaling in human neutrophils and monocytes. Human whole blood treated with the components of this SPM cluster enhanced both phagocytosis and killing of Escherichia coli by leukocytes. Thus, we identified a pro-resolving lipid mediator circuit and specific SPM cluster that promotes host defense. This new lipid mediator (LM)-SPM metabololipidomic approach now provides accessible metabolomic profiles in healthy and diseased human tissues, including cancer, for precision and personalized medicine.
Collapse
|
56
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
57
|
Chronic administration of mitochondrion-targeted peptide SS-31 prevents atherosclerotic development in ApoE knockout mice fed Western diet. PLoS One 2017; 12:e0185688. [PMID: 28961281 PMCID: PMC5621700 DOI: 10.1371/journal.pone.0185688] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/18/2017] [Indexed: 12/23/2022] Open
Abstract
Background Oxidative stress and inflammatory factors are deeply involved in progression of atherosclerosis. Mitochondrion-targeted peptide SS-31, selectively targeting to mitochondrial inner membrane reacting with cardiolipin, has been reported to inhibit ROS generation and mitigate inflammation. The present study was designed to investigate whether SS-31 could suppress the development of atherosclerosis in vivo. Methods Male ApoE-/- mice (8 weeks old) fed with Western diet were treated with normal saline or SS-31 (1 mg/kg/d or 3 mg/kg/d) through subcutaneous injection for 12 weeks. Oil Red O staining was performed to evaluate area and sizes of the plaques. DHE staining and immunohistochemical staining of 8-OHDG was performed to assess the oxidative stress. The aorta ATP contents were assessed by the ATP bioluminescence assay kit. Immunohistochemical staining of CD68 and α-SMA and Masson’s trichrome staining were performed to evaluate the composition of atherosclerotic plaque. Biochemical assays were performed to determine the protein level and activity of superoxide dismutase (SOD). The levels of CD36, LOX-1 and ABCA1 were immunohistochemically and biochemically determined to evaluate the cholesterol transport in aorta and peritoneal macrophages. Inflammatory factors, including ICAM-1, MCP-1, IL-6 and CRP in serum, were detected through ELISA. Results SS-31 administration reduced the area and sizes of western diet-induced atherosclerotic plaques and changed the composition of the plaques in ApoE-/- mice. Oxidative stress was suppressed, as evidenced by the reduced DHE stain, down-regulated 8-OHDG expression, and increased SOD activity after chronic SS-31 administration. Moreover, systemic inflammation was ameliorated as seen by decreasing serum ICAM-1, MCP-1, and IL-6 levels. Most importantly, SS-31 administration inhibited cholesterol influx by down-regulating expression of CD36 and LOX-1 to prevent lipid accumulation to further suppress the foam cell formation and atherosclerotic progression. Conclusion Administration of SS-31 prevents against atherosclerotic formation in ApoE-/- mice suggesting that SS-31 might be considered to be a potential drug to prevent atherosclerotic progression.
Collapse
|
58
|
Kassab SE, Khedr MA, Ali HI, Abdalla MM. Discovery of new indomethacin-based analogs with potentially selective cyclooxygenase-2 inhibition and observed diminishing to PGE2 activities. Eur J Med Chem 2017; 141:306-321. [PMID: 29031075 DOI: 10.1016/j.ejmech.2017.09.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 11/29/2022]
Abstract
New ring-extended analogs of indomethacin were designed based on the structure of active binding site of both COX-1 and COX-2 isoenzymes and the interaction pattern required for selective inhibition of COX-2 to improve its selectivity against COX-2. The strategy adopted for designing the new inhibitors involved i) ring extension of indomethacin to reduce the possibility of analogs to be accommodated into the narrow hydrophobic tunnel of COX-1, ii) deletion of carboxylic acid to reduce the possibility of inhibitor to form salt bridge with Arg120 and eventually prevent COX-1 inhibition, and iii) introduction of methylsulfonyl group to increase the opportunity of the analogs to interact with the polar side pocket that's is crucial for inhibition process of COX-2. The three series of tetrahydrocarbazoles involving 4, 5, 9, 10 and 12 were synthesized in quantitative yields adopting limited number of reaction steps, and applying laboratory friendly reaction conditions. In vitro and in vivo assays for data profiling the new candidates revealed the significant improvement in the potency and selectivity against COX-2 of 6-methoxytetrahydrocarbazole 4 (IC50 = 0.97 μmol) to verify the effect of ring extension in comparison to indomethacin (IC50 = 2.63 μmol), and 6-methylsulfonyltetrahydrocarbazole 10a (IC50 = 0.28 μmol) to verify the effect of ring extension and introduction of methylsulfonyl group. 9-(4-chlorobenzoyl)-6-(methylsulfonyl)-1,2,3,9-tetrahydro-4H-carbazol-4-one 12a showed the most potential and selective activity against COX-2 (IC50 = 0.23 μmol) to be with superior potency to Celecoxib (IC50 = 0.30 μmol). Consistently, 12a was the most active with all the other anti-inflammatory test descriptors and its activity in diminishing the PGE2 with the other analogs confirmed the elaboration of new class of selective COX-2 inhibitors beyond the diarylsulfonamides as a previously common class of selective COX-2 inhibitors. Molecular docking study revealed the high binding score of compound 12a (-30.78 kcal/mol), with less clash contribution (7.2) that is close to indomethacin. Also, 12a showed low conformation entropy score (1.40). Molecular dynamic (MD) simulation identified the equilibrium of both potential and kinetic energies.
Collapse
Affiliation(s)
- Shaymaa E Kassab
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt.
| | - Mohammed A Khedr
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Hamed I Ali
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; Department of Pharmaceutical Sciences, Texas A&M University Irma Lerma Rangel College of Pharmacy, Kingsville 78363, Texas, USA
| | - Mohamed M Abdalla
- Research Unit, Saco Pharm. Co., 6th of October City, Giza 68330, Egypt
| |
Collapse
|
59
|
Martinez JE, Taipeiro EDF, Chies AB. Effects of Continuous and Accumulated Exercise on Endothelial Function in Rat Aorta. Arq Bras Cardiol 2017; 108:315-322. [PMID: 28538761 PMCID: PMC5421470 DOI: 10.5935/abc.20170036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 11/21/2016] [Indexed: 12/29/2022] Open
Abstract
Background: The practice of exercise in short bouts repeated throughout the day may be an alternative strategy to lift people out of physical inactivity. Objective: to evaluate if accumulated exercise, as occurs in continuous exercise training, improve endothelial function in rat aorta. Methods: Wistar male rats were divided into three groups: continuous exercise (CEx, 1 hour on the treadmill) or accumulated exercise (AEx, 4 bouts of 15 minutes / day) for 5 days/week for 8 weeks, or sedentary (SED). During the training period, body weight gain and increase in exercise performance were recorded. On sacrifice day, aorta was dissected into rings (3-5 mm) and mounted on the organ bath. Results: Fitness was significantly greater in CEx and AEx rats as compared with SED animals. In addition, compared with the SED group, CEx animals had a lower body mass gain, and the aorta obtained from these animals had reduced contractile response to norepinephrine and greater acetylcholine-induced relaxation. These results were not observed in ACEx animals. Conclusions: Both CEx and AEx improved fitness, but only CEx led to reduced body weight gain and improved endothelial function. Fundamento: A prática de exercícios em sessões curtas que se repetem ao longo do dia pode ser uma alternativa para tirar as pessoas da inatividade física. Objetivo: Verificar se o exercício acumulado, tal como ocorre com o treinamento com exercício contínuo, melhora a função endotelial na aorta de ratos. Métodos: Ratos Wistar machos foram divididos em 3 grupos: treinamento com exercício contínuo (ExC; 1 hora em esteira) ou com exercício acumulado (ExA; 4 sessões de 15 minutos ao longo do dia) por 5 dias/semana, durante 8 semanas, ou grupo sedentário (SED). Durante o treinamento, foram registrados o ganho de peso corporal e desempenho na esteira. No dia do sacrifício, anéis (3-5 mm) da aorta foram obtidos e montados em banho de órgãos. Resultados: Animais ExC e ExA mostraram aptidão física significativamente maior em comparação com os SED. Paralelamente, em comparação com SED, animais ExC tiveram menor ganho de massa corporal, e aortas retiradas desses animais mostraram respostas contrácteis à noradrenalina reduzidas e maior relaxamento induzido pela acetilcolina. Esses resultados não foram observados no grupo ExA. Conclusões: Tanto o ExC quanto o ExA melhoraram a aptidão física, mas somente o ExC foi capaz de reduzir o ganho de peso corporal dos animais e melhorar a função endotelial.
Collapse
|
60
|
Norris PC, Libreros S, Chiang N, Serhan CN. A cluster of immunoresolvents links coagulation to innate host defense in human blood. Sci Signal 2017; 10:10/490/eaan1471. [PMID: 28765512 DOI: 10.1126/scisignal.aan1471] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Blood coagulation is a protective response that prevents excessive bleeding upon blood vessel injury. We investigated the relationship between coagulation and the resolution of inflammation and infection by lipid mediators (LMs) through metabololipidomics-based profiling of human whole blood (WB) during coagulation. We identified temporal clusters of endogenously produced prothrombotic and proinflammatory LMs (eicosanoids), as well as specialized proresolving mediators (SPMs). In addition to eicosanoids, a specific SPM cluster was identified that consisted of resolvin E1 (RvE1), RvD1, RvD5, lipoxin B4, and maresin 1, each of which was present at bioactive concentrations (0.1 to 1 nM). Removal of adenosine from the coagulating blood markedly enhanced the amounts of SPMs produced and further increased the biosynthesis of RvD3, RvD4, and RvD6. The cyclooxygenase inhibitors celecoxib and indomethacin, which block the production of thromboxanes and prostanoids, did not block the production of clot-driven SPMs. Unbiased mass cytometry analysis demonstrated that the SPM cluster produced in human blood targeted leukocytes at the single-cell level, directly activating ERK and CREB signaling in neutrophils and CD14+ monocytes. Treatment of human WB with the components of this SPM cluster enhanced both the phagocytosis and killing of Escherichia coli by leukocytes. Together, these data identify a proresolving LM circuit, including endogenous molecular brakes and accelerators, which promoted host defense. These temporal LM-SPM clusters can provide accessible metabolomic profiles for precision and personalized medicine.
Collapse
Affiliation(s)
- Paul C Norris
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephania Libreros
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nan Chiang
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
61
|
Grosser T, Ricciotti E, FitzGerald GA. The Cardiovascular Pharmacology of Nonsteroidal Anti-Inflammatory Drugs. Trends Pharmacol Sci 2017; 38:733-748. [PMID: 28651847 DOI: 10.1016/j.tips.2017.05.008] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/24/2017] [Accepted: 05/25/2017] [Indexed: 12/27/2022]
Abstract
The principal molecular mechanisms underlying the cardiovascular (CV) and renal adverse effects of nonsteroidal anti-inflammatory drugs (NSAIDs), such as myocardial infarction and hypertension, are understood in more detail than most side effects of drugs. Less is known, however, about differences in the CV safety profile between chemically distinct NSAIDs and their relative predisposition to complications. In review article, we discuss how heterogeneity in the pharmacokinetics and pharmacodynamics of distinct NSAIDs may be expected to affect their CV risk profile. We consider evidence afforded by studies in model systems, mechanistic clinical trials, a meta-analysis of randomized controlled trials, and two recent large clinical trials, Standard Care vs. Celecoxib Outcome Trial (SCOT) and Prospective Randomized Evaluation of Celecoxib Integrated Safety versus Ibuprofen or Naproxen (PRECISION), designed specifically to compare the CV safety of the cyclooxygenase-2-selective NSAID, celecoxib, with traditional NSAIDs. We conclude that SCOT and PRECISION have apparently not compared equipotent doses and have other limitations that bias them toward underestimation of the relative risk of celecoxib.
Collapse
Affiliation(s)
- Tilo Grosser
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emanuela Ricciotti
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
62
|
Tacconelli S, Bruno A, Grande R, Ballerini P, Patrignani P. Nonsteroidal anti-inflammatory drugs and cardiovascular safety – translating pharmacological data into clinical readouts. Expert Opin Drug Saf 2017; 16:791-807. [PMID: 28569569 DOI: 10.1080/14740338.2017.1338272] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Stefania Tacconelli
- Department of Neuroscience, Imaging and Clinical Sciences, ‘G. d’Annunzio’ University, Chieti, Italy
- Center for Aging and Translational Medicine (CeSI-MeT), ‘G. d’Annunzio’ University, Chieti, Italy
| | - Annalisa Bruno
- Department of Neuroscience, Imaging and Clinical Sciences, ‘G. d’Annunzio’ University, Chieti, Italy
- Center for Aging and Translational Medicine (CeSI-MeT), ‘G. d’Annunzio’ University, Chieti, Italy
| | - Rosalia Grande
- Department of Neuroscience, Imaging and Clinical Sciences, ‘G. d’Annunzio’ University, Chieti, Italy
- Center for Aging and Translational Medicine (CeSI-MeT), ‘G. d’Annunzio’ University, Chieti, Italy
| | - Patrizia Ballerini
- Center for Aging and Translational Medicine (CeSI-MeT), ‘G. d’Annunzio’ University, Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, ‘G.d’Annunzio’ University, Chieti, Italy
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences, ‘G. d’Annunzio’ University, Chieti, Italy
- Center for Aging and Translational Medicine (CeSI-MeT), ‘G. d’Annunzio’ University, Chieti, Italy
| |
Collapse
|
63
|
Oxygenated hypothermic machine perfusion after static cold storage improves endothelial function of extended criteria donor livers. HPB (Oxford) 2017; 19:538-546. [PMID: 28351756 DOI: 10.1016/j.hpb.2017.02.439] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/22/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lack of oxygen and biomechanical stimulation during static cold storage (SCS) of donor livers compromises endothelial cell function. We investigated the effect of end-ischemic oxygenated hypothermic machine perfusion (HMP) on endothelial cell function of extended criteria donor (ECD) livers. METHODS Eighteen livers, declined for transplantation, were transported to our center using static cold storage (SCS). After SCS, 6 livers underwent two hours of HMP, and subsequent normothermic machine perfusion (NMP) to assess viability. Twelve control livers underwent NMP immediately after SCS. mRNA expression of transcription factor Krüppel-like-factor 2 (KLF2), endothelial nitric oxide synthase (eNOS), and thrombomodulin (TM) was quantified by RT-PCR. Endothelial cell function and injury were assessed by nitric oxide (NO) production and release of TM into the perfusate. RESULTS In HMP livers, mRNA expression of KLF2 (p = 0.043), eNOS (p = 0.028), and TM (p = 0.028) increased significantly during NMP. In parallel, NO levels increased during NMP in HMP livers but not in controls. At the end of NMP cumulative TM release was significantly lower HMP livers, compared to controls (p = 0.028). CONCLUSION A short period of two hours oxygenated HMP restores endothelial cell viability after SCS and subsequent normothermic reoxygenation of ECD livers.
Collapse
|
64
|
Erbeldinger N, Rapp F, Ktitareva S, Wendel P, Bothe AS, Dettmering T, Durante M, Friedrich T, Bertulat B, Meyer S, Cardoso MC, Hehlgans S, Rödel F, Fournier C. Measuring Leukocyte Adhesion to (Primary) Endothelial Cells after Photon and Charged Particle Exposure with a Dedicated Laminar Flow Chamber. Front Immunol 2017; 8:627. [PMID: 28620384 PMCID: PMC5451490 DOI: 10.3389/fimmu.2017.00627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium interacts with all types of blood cells and is a key modulator of local and systemic inflammatory processes, for example, in the adhesion of blood leukocytes to endothelial cells (EC) and the following extravasation into the injured tissue. The endothelium is constantly exposed to mechanical forces caused by blood flow, and the resulting shear stress is essential for the maintenance of endothelial function. Changes in local hemodynamics are sensed by EC, leading to acute or persistent changes. Therefore, in vitro assessment of EC functionality should include shear stress as an essential parameter. Parallel-plate flow chambers with adjustable shear stress can be used to study EC properties. However, commercially available systems are not suitable for radiation experiments, especially with charged particles, which are increasingly used in radiotherapy of tumors. Therefore, research on charged-particle-induced vascular side effects is needed. In addition, α-particle emitters (e.g., radon) are used to treat inflammatory diseases at low doses. In the present study, we established a flow chamber system, applicable for the investigation of radiation induced changes in the adhesion of lymphocytes to EC as readout for the onset of an inflammatory reaction or the modification of a pre-existing inflammatory state. In this system, primary human EC are cultured under physiological laminar shear stress, subjected to a proinflammatory treatment and/or irradiation with X-rays or charged particles, followed by a coincubation with primary human lymphocytes (peripheral blood lymphocytes (PBL)). Analysis is performed by semiautomated quantification of fluorescent staining in microscopic pictures. First results obtained after irradiation with X-rays or helium ions indicate decreased adhesion of PBL to EC under laminar conditions for both radiation qualities, whereas adhesion of PBL under static conditions is not clearly affected by irradiation. Under static conditions, no radiation-induced changes in surface expression of adhesion molecules and activation of nuclear factor kappa B (NF-κB) signaling were observed after single cell-based high-throughput analysis. In subsequent studies, these investigations will be extended to laminar conditions.
Collapse
Affiliation(s)
- Nadine Erbeldinger
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany.,Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - Felicitas Rapp
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Svetlana Ktitareva
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Philipp Wendel
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Anna S Bothe
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Till Dettmering
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Marco Durante
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Thomas Friedrich
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Bianca Bertulat
- Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - Stephanie Meyer
- Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - M C Cardoso
- Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, University of Frankfurt, Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, University of Frankfurt, Frankfurt, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| |
Collapse
|
65
|
Patrono C, Baigent C. Coxibs, Traditional NSAIDs, and Cardiovascular Safety Post-PRECISION: What We Thought We Knew Then and What We Think We Know Now. Clin Pharmacol Ther 2017; 102:238-245. [PMID: 28378879 DOI: 10.1002/cpt.696] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/15/2017] [Indexed: 01/08/2023]
Abstract
The aim of the present review is to analyze how thinking about the cardiovascular safety of nonsteroidal antiinflammatory drugs has evolved during the past two decades, and discuss to what extent the additional information from the Prospective Randomized Evaluation of Celecoxib Integrated Safety Versus Ibuprofen or Naproxen study may alter our current mechanistic understanding and/or clinical practice.
Collapse
Affiliation(s)
- C Patrono
- Department of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - C Baigent
- Medical Research Council Population Health Research Unit, and Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
66
|
Diaz MF, Vaidya AB, Evans SM, Lee HJ, Aertker BM, Alexander AJ, Price KM, Ozuna JA, Liao GP, Aroom KR, Xue H, Gu L, Omichi R, Bedi S, Olson SD, Cox CS, Wenzel PL. Biomechanical Forces Promote Immune Regulatory Function of Bone Marrow Mesenchymal Stromal Cells. Stem Cells 2017; 35:1259-1272. [PMID: 28181347 PMCID: PMC5405000 DOI: 10.1002/stem.2587] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/17/2016] [Accepted: 01/24/2017] [Indexed: 01/03/2023]
Abstract
Mesenchymal stromal cells (MSCs) are believed to mobilize from the bone marrow in response to inflammation and injury, yet the effects of egress into the vasculature on MSC function are largely unknown. Here we show that wall shear stress (WSS) typical of fluid frictional forces present on the vascular lumen stimulates antioxidant and anti-inflammatory mediators, as well as chemokines capable of immune cell recruitment. WSS specifically promotes signaling through NFκB-COX2-prostaglandin E2 (PGE2 ) to suppress tumor necrosis factor-α (TNF-α) production by activated immune cells. Ex vivo conditioning of MSCs by WSS improved therapeutic efficacy in a rat model of traumatic brain injury, as evidenced by decreased apoptotic and M1-type activated microglia in the hippocampus. These results demonstrate that force provides critical cues to MSCs residing at the vascular interface which influence immunomodulatory and paracrine activity, and suggest the potential therapeutic use of force for MSC functional enhancement. Stem Cells 2017;35:1259-1272.
Collapse
Affiliation(s)
- Miguel F. Diaz
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Abishek B. Vaidya
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Siobahn M. Evans
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Hyun J. Lee
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Benjamin M. Aertker
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Alexander J. Alexander
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
- Department of BioSciences, Rice University, Houston, TX 77030, USA
| | - Katherine M. Price
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
- Department of BioSciences, Rice University, Houston, TX 77030, USA
| | - Joyce A. Ozuna
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
- Department of BioSciences, Rice University, Houston, TX 77030, USA
| | - George P. Liao
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Kevin R. Aroom
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Hasen Xue
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Liang Gu
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Rui Omichi
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
- School of Medicine, Faculty of Medicine, Tokushima University, Tokushima 770-8501, Japan
| | - Supinder Bedi
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Scott D. Olson
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Charles S. Cox
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| | - Pamela L. Wenzel
- Children’s Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, TX, 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, 77030, USA
| |
Collapse
|
67
|
Bravo B, García de Durango C, González Á, Gortázar AR, Santos X, Forteza-Vila J, Vidal-Vanaclocha F. Opposite Effects of Mechanical Action of Fluid Flow on Proangiogenic Factor Secretion From Human Adipose-Derived Stem Cells With and Without Oxidative Stress. J Cell Physiol 2017; 232:2158-2167. [PMID: 27925206 DOI: 10.1002/jcp.25712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/29/2016] [Indexed: 12/30/2022]
Abstract
Mechanical forces, hypoxia, and oxidative stress contribute to skin renewal, perfusion, and wound healing, but how are they regulating subcutaneous adipose-derived stem cells (ASCs) in the inflammatory microenvironment associated to skin repair and disorders is unknown. In this study, ASCs were isolated from lipoaspirate samples from plastic surgery patients, primary cultured and their differentiation and secretion of a panel of cytokines with pronounced effects on skin repair and angiogenesis were studied under mechanical stimulation by intermittent fluid flow, 1% hypoxia and oxidative stress by glutathione (GSH) depletion with buthionine sulfoximine (BSO) treatment. Mechanical action of fluid flow did not alter mesenchymal phenotype of CD90+ /CD29+ /CD44+ /CD34- /CD106- /CD45- ASCs; however, it remarkably induced ASC secretion of human umbilical vein endothelial cell (HUVEC) migration-stimulating factors. Multiplex Luminex assay further confirmed an increased secretion of VEGF, G-CSF, HGF, Leptin, IL-8, PDGF-BB, Angiopoietin-2, and Follistatin from mechanically-stimulated ASCs via cyclooxygenase-2. Consistent with this mechanism, GSH depletion and hypoxia also increased ASC secretion of VEGF, IL-8, leptin, Angiopoitein-2, and PDGF-BB. However, mechanical action of fluid flow abrogated VEGF and HUVEC migration-stimulating activity from GSH-depleted and hypoxic ASCs. Conversely, GSH depletion and hypoxia abrogated VEGF and HUVEC migration-stimulating activity from mechano-stimulated ASCs. Although mechanical action of fluid flow, hypoxia, and GSH-depletion had independent proangiogenic-stimulating activity on ASCs, mechanical stimulation had opposite effects on proangiogenic factor secretion from ASCs with and without oxidative stress. These data uncover the role of hypoxia and endogenous redox balance during the proangiogenic response of ASCs and other mesenchymal-derived cell types to mechanical action of interstitial fluid flow. J. Cell. Physiol. 232: 2158-2167, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Beatriz Bravo
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | - Cira García de Durango
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | - Álvaro González
- Department of Molecular and Cellular Oncology Houston, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arancha R Gortázar
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | - Xavier Santos
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | - Jerónimo Forteza-Vila
- Valencia Institute of Pathology (IVP), Catholic University of Valencia School of Medicine and Odontology, Valencia, Spain
| | - Fernando Vidal-Vanaclocha
- Valencia Institute of Pathology (IVP), Catholic University of Valencia School of Medicine and Odontology, Valencia, Spain
| |
Collapse
|
68
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 635] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
69
|
Di Russo J, Luik AL, Yousif L, Budny S, Oberleithner H, Hofschröer V, Klingauf J, van Bavel E, Bakker EN, Hellstrand P, Bhattachariya A, Albinsson S, Pincet F, Hallmann R, Sorokin LM. Endothelial basement membrane laminin 511 is essential for shear stress response. EMBO J 2016; 36:183-201. [PMID: 27940654 PMCID: PMC5239996 DOI: 10.15252/embj.201694756] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 11/09/2022] Open
Abstract
Shear detection and mechanotransduction by arterial endothelium requires junctional complexes containing PECAM-1 and VE-cadherin, as well as firm anchorage to the underlying basement membrane. While considerable information is available for junctional complexes in these processes, gained largely from in vitro studies, little is known about the contribution of the endothelial basement membrane. Using resistance artery explants, we show that the integral endothelial basement membrane component, laminin 511 (laminin α5), is central to shear detection and mechanotransduction and its elimination at this site results in ablation of dilation in response to increased shear stress. Loss of endothelial laminin 511 correlates with reduced cortical stiffness of arterial endothelium in vivo, smaller integrin β1-positive/vinculin-positive focal adhesions, and reduced junctional association of actin-myosin II In vitro assays reveal that β1 integrin-mediated interaction with laminin 511 results in high strengths of adhesion, which promotes p120 catenin association with VE-cadherin, stabilizing it at cell junctions and increasing cell-cell adhesion strength. This highlights the importance of endothelial laminin 511 in shear response in the physiologically relevant context of resistance arteries.
Collapse
Affiliation(s)
- Jacopo Di Russo
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Anna-Liisa Luik
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Lema Yousif
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Sigmund Budny
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Hans Oberleithner
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Verena Hofschröer
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Juergen Klingauf
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Medical Physics, University of Muenster, Muenster, Germany
| | - Ed van Bavel
- Biomedical Engineering and Physics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Ntp Bakker
- Biomedical Engineering and Physics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Frederic Pincet
- Laboratoire de Physique Statistique, École Normale Superieure - PSL Research University, Paris, France.,CNRS UMR8550, Sorbonne Universités - UPMC Univ Paris 06, Université Paris, Paris, France
| | - Rupert Hallmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Lydia M Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany .,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| |
Collapse
|
70
|
Egginton S, Hussain A, Hall-Jones J, Chaudhry B, Syeda F, Glen KE. Shear stress-induced angiogenesis in mouse muscle is independent of the vasodilator mechanism and quickly reversible. Acta Physiol (Oxf) 2016; 218:153-166. [PMID: 27261201 PMCID: PMC5082534 DOI: 10.1111/apha.12728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/10/2015] [Accepted: 06/01/2016] [Indexed: 11/29/2022]
Abstract
AIM Is modulation of skeletal muscle capillary supply by altering blood flow due to a presumptive shear stress response per se, or dependent on the vasodilator mechanism? METHODS The response to four different vasodilators, and cotreatment with blockers of NO and prostaglandin synthesis, was compared. Femoral artery blood flow was correlated with capillary-to-fibre ratio (C:F) and protein levels of putative angiogenic compounds. RESULTS All vasodilators induced a similar increase in blood flow after 14 days, with a similar effect on C:F (1.62 ± 0.05, 1.60 ± 0.01, 1.57 ± 0.06, 1.57 ± 0.07, respectively, all P < 0.05 vs. control 1.20 ± 0.01). Concomitant inhibitors revealed differential effects on blood flow and angiogenesis, demonstrating that a similar response may have different signalling origins. The time course of this response with the most commonly used vasodilator, prazosin, showed that blood flow increased from 0.40 mL min-1 to 0.61 mL min-1 by 28 days (P < 0.05), dropped within 1 week after the cessation of treatment (0.54 mL min-1 ; P < 0.05) and returned to control levels by 6 weeks. In parallel with FBF, capillary rarefaction began within 1 week (P < 0.05), giving C:F values similar to control by 2 weeks. Of the dominant signalling pathways, prazosin decreased muscle VEGF, but increased its cognate receptor Flk-1 (both P < 0.01); levels of eNOS varied with blood flow (P < 0.05), and Ang-1 initially increased, while its receptor Tie-2 was unchanged, with only modest changes in the antiangiogenic factor TSP-1. CONCLUSION Hyperaemia-induced angiogenesis, likely in response to elevated shear stress, is independent of the vasodilator involved, with a rapid induction and quick regression following the stimulus withdrawal.
Collapse
Affiliation(s)
- S. Egginton
- School of Biomedical Sciences; University of Leeds; Leeds UK
| | - A. Hussain
- Science Department; Denefield School; Reading UK
- Centre for Cardiovascular Sciences; Medical School; University of Birmingham; Birmingham UK
| | - J. Hall-Jones
- Centre for Cardiovascular Sciences; Medical School; University of Birmingham; Birmingham UK
| | - B. Chaudhry
- Centre for Cardiovascular Sciences; Medical School; University of Birmingham; Birmingham UK
| | - F. Syeda
- Centre for Cardiovascular Sciences; Medical School; University of Birmingham; Birmingham UK
| | - K. E. Glen
- Centre for Biological Engineering; Loughborough University; Loughborough UK
| |
Collapse
|
71
|
Cancel LM, Ebong EE, Mensah S, Hirschberg C, Tarbell JM. Endothelial glycocalyx, apoptosis and inflammation in an atherosclerotic mouse model. Atherosclerosis 2016; 252:136-146. [PMID: 27529818 DOI: 10.1016/j.atherosclerosis.2016.07.930] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/20/2016] [Accepted: 07/28/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Previous experiments suggest that both increased endothelial cell apoptosis and endothelial surface glycocalyx shedding could play a role in the endothelial dysfunction and inflammation of athero-prone regions of the vasculature. We sought to elucidate the possibly synergistic mechanisms by which endothelial cell apoptosis and glycocalyx shedding promote atherogenesis. METHODS 4- to 6-week old male C57Bl/6 apolipoprotein E knockout (ApoE(-/-)) mice were fed a Western diet for 10 weeks and developed plaques in their brachiocephalic arteries. RESULTS Glycocalyx coverage and thickness were significantly reduced over the plaque region compared to the non-plaque region (coverage plaque: 71 ± 23%, non-plaque: 97 ± 3%, p = 0.02; thickness plaque: 0.85 ± 0.15 μm, non-plaque: 1.2 ± 0.21 μm, p = 0.006). Values in the non-plaque region were not different from those found in wild type mice fed a normal diet (coverage WT: 92 ± 3%, p = 0.7 vs. non-plaque ApoE(-/-), thickness WT: 1.1 ± 0.06 μm, p = 0.2 vs. non-plaque ApoE(-/-)). Endothelial cell apoptosis was significantly increased in ApoE(-/-) mice compared to wild type mice (ApoE(-/-):64.3 ± 33.0, WT: 1.1 ± 0.5 TUNEL-pos/cm, p = 2 × 10(-7)). The number of apoptotic endothelial cells per unit length was 2 times higher in the plaque region than in the non-plaque region of the same vessel (p = 3 × 10(-5)). Increased expression of matrix metalloproteinase 9 co-localized with glycocalyx shedding and plaque buildup. CONCLUSIONS Our results suggest that, in concert with endothelial apoptosis that increases lipid permeability, glycocalyx shedding initiated by inflammation facilitates monocyte adhesion and macrophage infiltration that promote lipid retention and the development of atherosclerotic plaques.
Collapse
Affiliation(s)
- Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| | - Eno E Ebong
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Solomon Mensah
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Carly Hirschberg
- Graduate Division Summer Undergraduate Research Program, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
72
|
Fanaroff AC, Roe MT. Contemporary Reflections on the Safety of Long-Term Aspirin Treatment for the Secondary Prevention of Cardiovascular Disease. Drug Saf 2016; 39:715-27. [PMID: 27028617 PMCID: PMC5778440 DOI: 10.1007/s40264-016-0421-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aspirin has been the cornerstone of therapy for the secondary prevention treatment of patients with cardiovascular disease since landmark trials were completed in the late 1970s and early 1980s that demonstrated the efficacy of aspirin for reducing the risk of ischemic events. Notwithstanding the consistent benefits demonstrated with aspirin for both acute and chronic cardiovascular disease, there are a number of toxicities associated with aspirin that have been showcased by recent long-term clinical trials that have included an aspirin monotherapy arm. As an inhibitor of cyclooxygenase (COX), aspirin impairs gastric mucosal protective mechanisms. Previous trials have shown that up to 15-20 % of patients developed gastrointestinal symptoms with aspirin monotherapy, and approximately 1 % of patients per year had a clinically significant bleeding event, including 1 in 1000 patients who suffered an intracranial or fatal bleed. These risks have been shown to be compounded for patients with acute coronary syndromes (ACS) and those undergoing percutaneous coronary intervention (PCI) who are also treated with other antithrombotic agents during the acute care/procedural period, as well as for an extended time period afterwards. Given observations of substantial increases in bleeding rates from many prior long-term clinical trials that have evaluated aspirin together with other oral platelet inhibitors or oral anticoagulants, the focus of contemporary research has pivoted towards tailored antithrombotic regimens that attempt to either shorten the duration of exposure to aspirin or replace aspirin with an alternative antithrombotic agent. While these shifts are occurring, the safety profile of aspirin when used for the secondary prevention treatment of patients with established cardiovascular disease deserves further consideration.
Collapse
Affiliation(s)
- Alexander C Fanaroff
- Duke Clinical Research Institute, Duke University Medical Center, 2400 Pratt Street, Room 7035, Durham, NC, 27705, USA
| | - Matthew T Roe
- Duke Clinical Research Institute, Duke University Medical Center, 2400 Pratt Street, Room 7035, Durham, NC, 27705, USA.
| |
Collapse
|
73
|
Qiao C, Meng F, Jang I, Jo H, Chen YE, Zhang J. Deep transcriptomic profiling reveals the similarity between endothelial cells cultured under static and oscillatory shear stress conditions. Physiol Genomics 2016; 48:660-6. [PMID: 27449656 DOI: 10.1152/physiolgenomics.00025.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/17/2016] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis is a multifactorial disease that preferentially develops in specific regions in the arterial tree. This characteristic is mainly attributed to the unique pattern of hemodynamic shear stress in vivo. High laminar shear stress (LS) found in straight lumen exerts athero-protective effects. Low or oscillatory shear stress (OS) present in regions of lesser curvature and arterial bifurcations predisposes arterial intima to atherosclerosis. Shear stress-regulated endothelial function plays an important role in the process of atherosclerosis. Most in vitro research studies focusing on the molecular mechanisms of endothelial function are performed in endothelial cells (ECs) under cultured static (ST) condition. Some findings, however, are not recapitulated in subsequent translational studies, mostly likely due to the missing biomechanical milieu. Here, we profiled the whole transcriptome of primary human coronary arterial endothelial cells (HCAECs) under different shear stress conditions with RNA sequencing. Among 16,313 well-expressed genes, we detected 8,177 that were differentially expressed in OS vs. LS conditions and 9,369 in ST vs. LS conditions. Notably, only 1,618 were differentially expressed in OS vs. ST conditions. Hierarchical clustering of ECs demonstrated a strong similarity between ECs under OS and ST conditions at the transcriptome level. Subsequent pairwise heat mapping and principal component analysis gave further weight to the similarity. At the individual gene level, expressional analysis of representative well-known genes as well as novel genes showed a comparable amount at mRNA and protein levels in ECs under ST and OS conditions. In conclusion, the present work compared the whole transcriptome of HCAECs under different shear stress conditions at the transcriptome level as well as at the individual gene level. We found that cultured ECs are significantly different from those under LS conditions. Thus using cells under ST conditions is unlikely to elucidate endothelial physiology. Given the revealed high similarities of the endothelial transcriptome under OS and ST conditions, it may be helpful to understand the underlying mechanisms of OS-induced endothelial dysfunction from static cultured endothelial studies.
Collapse
Affiliation(s)
- Congzhen Qiao
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Fan Meng
- Department of Psychiatry and Molecular and Behavioral Neuroscience Institute, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - Inhwan Jang
- Wallace H. Coulter Department of Biomedical Engineering, Division of Cardiology, Department of Medicine, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Division of Cardiology, Department of Medicine, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Y Eugene Chen
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Jifeng Zhang
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan;
| |
Collapse
|
74
|
Abstract
Dysfunction of the endothelial lining of lesion-prone areas of the arterial vasculature is an important contributor to the pathobiology of atherosclerotic cardiovascular disease. Endothelial cell dysfunction, in its broadest sense, encompasses a constellation of various nonadaptive alterations in functional phenotype, which have important implications for the regulation of hemostasis and thrombosis, local vascular tone and redox balance, and the orchestration of acute and chronic inflammatory reactions within the arterial wall. In this review, we trace the evolution of the concept of endothelial cell dysfunction, focusing on recent insights into the cellular and molecular mechanisms that underlie its pivotal roles in atherosclerotic lesion initiation and progression; explore its relationship to classic, as well as more recently defined, clinical risk factors for atherosclerotic cardiovascular disease; consider current approaches to the clinical assessment of endothelial cell dysfunction; and outline some promising new directions for its early detection and treatment.
Collapse
Affiliation(s)
- Michael A Gimbrone
- From the Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| | - Guillermo García-Cardeña
- From the Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
75
|
Simmons RD, Kumar S, Thabet SR, Sur S, Jo H. Omics-based approaches to understand mechanosensitive endothelial biology and atherosclerosis. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:378-401. [PMID: 27341633 DOI: 10.1002/wsbm.1344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is a multifactorial disease that preferentially occurs in arterial regions exposed to d-flow can be used to indicate disturbed flow or disturbed blood flow. The mechanisms by which d-flow induces atherosclerosis involve changes in the transcriptome, methylome, proteome, and metabolome of multiple vascular cells, especially endothelial cells. Initially, we begin with the pathogenesis of atherosclerosis and the changes that occur at multiple levels owing to d-flow, especially in the endothelium. Also, there are a variety of strategies used for the global profiling of the genome, transcriptome, miRNA-ome, DNA methylome, and metabolome that are important to define the biological and pathophysiological mechanisms of endothelial dysfunction and atherosclerosis. Finally, systems biology can be used to integrate these 'omics' datasets, especially those that derive data based on a single animal model, in order to better understand the pathophysiology of atherosclerosis development in a holistic manner and how this integrative approach could be used to identify novel molecular diagnostics and therapeutic targets to prevent or treat atherosclerosis. WIREs Syst Biol Med 2016, 8:378-401. doi: 10.1002/wsbm.1344 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Rachel D Simmons
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandeep Kumar
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Salim Raid Thabet
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sanjoli Sur
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
76
|
Scholz B, Korn C, Wojtarowicz J, Mogler C, Augustin I, Boutros M, Niehrs C, Augustin HG. Endothelial RSPO3 Controls Vascular Stability and Pruning through Non-canonical WNT/Ca(2+)/NFAT Signaling. Dev Cell 2016; 36:79-93. [PMID: 26766444 DOI: 10.1016/j.devcel.2015.12.015] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 11/16/2015] [Accepted: 12/11/2015] [Indexed: 12/15/2022]
Abstract
The WNT signaling enhancer R-spondin3 (RSPO3) is prominently expressed in the vasculature. Correspondingly, embryonic lethality of Rspo3-deficient mice is caused by vessel remodeling defects. Yet the mechanisms underlying vascular RSPO3 function remain elusive. Inducible endothelial Rspo3 deletion (Rspo3-iECKO) resulted in perturbed developmental and tumor vascular remodeling. Endothelial cell apoptosis and vascular pruning led to reduced microvessel density in Rspo3-iECKO mice. Rspo3-iECKO mice strikingly phenocopied the non-canonical WNT signaling-induced vascular defects of mice deleted for the WNT secretion factor Evi/Wls. An endothelial screen for RSPO3 and EVI/WLS co-regulated genes identified Rnf213, Usp18, and Trim30α. RNF213 targets filamin A and NFAT1 for proteasomal degradation attenuating non-canonical WNT/Ca(2+) signaling. Likewise, USP18 and TRIM5α inhibited NFAT1 activation. Consequently, NFAT protein levels were decreased in endothelial cells of Rspo3-iECKO mice and pharmacological NFAT inhibition phenocopied Rspo3-iECKO mice. The data identify endothelial RSPO3-driven non-canonical WNT/Ca(2+)/NFAT signaling as a critical maintenance pathway of the remodeling vasculature.
Collapse
Affiliation(s)
- Beate Scholz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Claudia Korn
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jessica Wojtarowicz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Carolin Mogler
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Iris Augustin
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; DNA Demethylation, DNA Repair and Reprogramming, Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; German Cancer Consortium, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
77
|
Zhang H, Liu J, Qu D, Wang L, Luo JY, Lau CW, Liu P, Gao Z, Tipoe GL, Lee HK, Ng CF, Ma RCW, Yao X, Huang Y. Inhibition of miR-200c Restores Endothelial Function in Diabetic Mice Through Suppression of COX-2. Diabetes 2016; 65:1196-207. [PMID: 26822089 DOI: 10.2337/db15-1067] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/06/2016] [Indexed: 11/13/2022]
Abstract
Endothelial dysfunction plays a crucial role in the development of diabetic vasculopathy. Our initial quantitative PCR results showed an increased miR-200c expression in arteries from diabetic mice and patients with diabetes. However, whether miR-200c is involved in diabetic endothelial dysfunction is unknown. Overexpression of miR-200c impaired endothelium-dependent relaxations (EDRs) in nondiabetic mouse aortas, whereas suppression of miR-200c by anti-miR-200c enhanced EDRs in diabetic db/db mice. miR-200c suppressed ZEB1 expression, and ZEB1 overexpression ameliorated endothelial dysfunction induced by miR-200c or associated with diabetes. More importantly, overexpression of anti-miR-200c or ZEB1 in vivo attenuated miR-200c expression and improved EDRs in db/db mice. Mechanistic study with the use of COX-2(-/-) mice revealed that COX-2 mediated miR-200c-induced endothelial dysfunction and that miR-200c upregulated COX-2 expression in endothelial cells through suppression of ZEB1 and increased production of prostaglandin E2, which also reduced EDR. This study demonstrates for the first time to our knowledge that miR-200c is a new mediator of diabetic endothelial dysfunction and inhibition of miR-200c rescues EDRs in diabetic mice. These new findings suggest the potential usefulness of miR-200c as the target for drug intervention against diabetic vascular complications.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Cell Line
- Cells, Cultured
- Cyclooxygenase 2/chemistry
- Cyclooxygenase 2/metabolism
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Diabetes Mellitus/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Gene Expression Regulation
- Humans
- In Vitro Techniques
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/metabolism
- Middle Aged
- RNA/metabolism
- RNA Interference
- Renal Artery/metabolism
- Renal Artery/pathology
- Renal Artery/physiopathology
- Vasodilation
- Zinc Finger E-box-Binding Homeobox 1/antagonists & inhibitors
- Zinc Finger E-box-Binding Homeobox 1/genetics
- Zinc Finger E-box-Binding Homeobox 1/metabolism
Collapse
Affiliation(s)
- Huina Zhang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jian Liu
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dan Qu
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang-Yun Luo
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhen Gao
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - George L Tipoe
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | - Hung Kay Lee
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiang Yao
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
78
|
Schmidt M, Lamberts M, Olsen AMS, Fosbøll E, Niessner A, Tamargo J, Rosano G, Agewall S, Kaski JC, Kjeldsen K, Lewis BS, Torp-Pedersen C. Cardiovascular safety of non-aspirin non-steroidal anti-inflammatory drugs: review and position paper by the working group for Cardiovascular Pharmacotherapy of the European Society of Cardiology. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2016; 2:108-18. [DOI: 10.1093/ehjcvp/pvv054] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/07/2015] [Indexed: 01/09/2023]
|
79
|
Schmidt M, Lamberts M, Olsen AMS, Fosbøll E, Niessner A, Tamargo J, Rosano G, Agewall S, Kaski JC, Kjeldsen K, Lewis BS, Torp-Pedersen C. Cardiovascular safety of non-aspirin non-steroidal anti-inflammatory drugs: review and position paper by the working group for Cardiovascular Pharmacotherapy of the European Society of Cardiology. Eur Heart J 2016; 37:1015-23. [DOI: 10.1093/eurheartj/ehv505] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/07/2015] [Indexed: 12/30/2022] Open
|
80
|
Thomason J, Lunsford K, Mackin A. Anti-platelet therapy in small animal medicine. J Vet Pharmacol Ther 2016; 39:318-35. [DOI: 10.1111/jvp.12301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 01/29/2016] [Indexed: 01/29/2023]
Affiliation(s)
- J. Thomason
- Department of Clinical Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| | - K. Lunsford
- Department of Clinical Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| | - A. Mackin
- Department of Clinical Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| |
Collapse
|
81
|
|
82
|
The role of endothelial mechanosensitive genes in atherosclerosis and omics approaches. Arch Biochem Biophys 2015; 591:111-31. [PMID: 26686737 DOI: 10.1016/j.abb.2015.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the U.S., and is a multifactorial disease that preferentially occurs in regions of the arterial tree exposed to disturbed blood flow. The detailed mechanisms by which d-flow induces atherosclerosis involve changes in the expression of genes, epigenetic patterns, and metabolites of multiple vascular cells, especially endothelial cells. This review presents an overview of endothelial mechanobiology and its relation to the pathogenesis of atherosclerosis with special reference to the anatomy of the artery and the underlying fluid mechanics, followed by a discussion of a variety of experimental models to study the role of fluid mechanics and atherosclerosis. Various in vitro and in vivo models to study the role of flow in endothelial biology and pathobiology are discussed in this review. Furthermore, strategies used for the global profiling of the genome, transcriptome, miR-nome, DNA methylome, and metabolome, as they are important to define the biological and pathophysiological mechanisms of atherosclerosis. These "omics" approaches, especially those which derive data based on a single animal model, provide unprecedented opportunities to not only better understand the pathophysiology of atherosclerosis development in a holistic and integrative manner, but also to identify novel molecular and diagnostic targets.
Collapse
|
83
|
Palma Zochio Tozzato G, Taipeiro EF, Spadella MA, Marabini Filho P, de Assis MR, Carlos CP, Girol AP, Chies AB. Collagen-induced arthritis increases inducible nitric oxide synthase not only in aorta but also in the cardiac and renal microcirculation of mice. Clin Exp Immunol 2015; 183:341-9. [PMID: 26456019 DOI: 10.1111/cei.12728] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 09/24/2015] [Accepted: 10/08/2015] [Indexed: 01/08/2023] Open
Abstract
Rheumatoid arthritis (RA) may promote endothelial dysfunction. This phenomenon requires further investigation, especially in collagen-induced arthritis (CIA), as it is considered the experimental model most similar to RA. The objectives of this study were to identify CIA-induced changes in noradrenaline (NE) and acetylcholine (ACh) responses in mice aortas that may suggest endothelial dysfunction in these animals. Moreover, we characterize CIA-induced modifications in inducible nitric oxide synthase (iNOS) expression in the aortas and cardiac and renal tissues taken from these mice that may be related to possible endothelial dysfunction. Male DBA/1J mice were immunized with 100 μg of emulsified bovine collagen type II (CII) plus complete Freund's adjuvant. Twenty-one days later, these animals received a boost of an additional 100 μg plus incomplete Freund's adjuvant. Fifteen days after the onset of the disease, aortic rings from CIA and control mice were challenged with NE and ACh in an organ bath. In these animals, iNOS was detected through immunohistochemical analysis of aorta, heart and kidneys. Plasma nitrite concentration was determined using the Griess reaction. CIA did not change NE or ACh responses in mice aorta but apparently increased the iNOS expression not only in aorta, but also in cardiac and renal microcirculation. In parallel, CIA reduced nitrite plasma concentration. In mice, CIA appears to increase the presence of iNOS in aorta, as well as in heart and in kidney microcirculation. This iNOS increase occurs apparently in parallel to a reduction of the bioavailability of NO. This phenomenon does not appear to change NE or ACh responses in aorta.
Collapse
Affiliation(s)
| | - E F Taipeiro
- Discipline of Biochemistry, Marília Medical School, Marília, SP, Brazil
| | - M A Spadella
- Discipline of Human Embryology, Marília Medical School, Marília, SP, Brazil
| | - P Marabini Filho
- Discipline of Pathology, Marília Medical School, Marília, SP, Brazil
| | - M R de Assis
- Discipline of Rheumatology, Marília Medical School, Marília, SP, Brazil
| | - C P Carlos
- Faceres School of Medicine, São José Do Rio Preto, SP, Brazil
| | - A P Girol
- Laboratory of Immunohistochemistry, Padre Albino School of Medicine, Catanduva, SP, Brazil
| | - A B Chies
- Laboratory of Pharmacology, Marília Medical School, Marília, SP, Brazil
| |
Collapse
|
84
|
Krishnan R, Park JA, Seow CY, Lee PVS, Stewart AG. Cellular Biomechanics in Drug Screening and Evaluation: Mechanopharmacology. Trends Pharmacol Sci 2015; 37:87-100. [PMID: 26651416 DOI: 10.1016/j.tips.2015.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/12/2015] [Accepted: 10/23/2015] [Indexed: 12/14/2022]
Abstract
The study of mechanobiology is now widespread. The impact of cell and tissue mechanics on cellular responses is well appreciated. However, knowledge of the impact of cell and tissue mechanics on pharmacological responsiveness, and its application to drug screening and mechanistic investigations, have been very limited in scope. We emphasize the need for a heightened awareness of the important bidirectional influence of drugs and biomechanics in all living systems. We propose that the term 'mechanopharmacology' be applied to approaches that employ in vitro systems, biomechanically appropriate to the relevant (patho)physiology, to identify new drugs and drug targets. This article describes the models and techniques that are being developed to transform drug screening and evaluation, ranging from a 2D environment to the dynamic 3D environment of the target expressed in the disease of interest.
Collapse
Affiliation(s)
- Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jin-Ah Park
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chun Y Seow
- Center for Heart Lung Innovation, St Pauls Hospital, University of British Columbia, Vancouver, Canada
| | - Peter V-S Lee
- Department of Mechanical Engineering, University of Melbourne, Melbourne, Australia
| | - Alastair G Stewart
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
85
|
Hasenberg T, Mühleder S, Dotzler A, Bauer S, Labuda K, Holnthoner W, Redl H, Lauster R, Marx U. Emulating human microcapillaries in a multi-organ-chip platform. J Biotechnol 2015; 216:1-10. [PMID: 26435219 DOI: 10.1016/j.jbiotec.2015.09.038] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023]
Abstract
Current microfluidic chip-based tissue culture systems lack a capillary endothelial vessel system, which would enable perfusion with blood. We utilise spatial cell cultures to populate a perfused multi-organ-chip platform-a microfluidic device recently introduced for substance testing. Complete biological vascularization of such culture systems is vital to properly emulate physiological tissue behaviour. In this study, we incorporated a fibrin scaffold into the two-organ-chip design. Herein, adipose-derived stromal cells (ASCs) directed human umbilical vein endothelial cells (HUVECs) to organise into tube-like structures. The ASCs induced tube formation of HUVECs in static and dynamic conditions. The replacement of full medium enriched with growth factors and foetal calf serum with basal medium resulted in viable cells with similar gene expression profiles. We regard this as a prerequisite for studies with organ constructs that have a need for a different medium formulation. Furthermore, we here address stability issues of the fibrin gel and fibrin composition for optimal microvessel formation.
Collapse
Affiliation(s)
- Tobias Hasenberg
- Technische Universität Berlin, Medical Biotechnology, TIB 4/4-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany; TissUse GmbH, Markgrafenstraße 18, 15528 Spreenhagen, Germany.
| | - Severin Mühleder
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Andrea Dotzler
- Technische Universität Berlin, Medical Biotechnology, TIB 4/4-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany; TissUse GmbH, Markgrafenstraße 18, 15528 Spreenhagen, Germany.
| | - Sophie Bauer
- Technische Universität Berlin, Medical Biotechnology, TIB 4/4-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Krystyna Labuda
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Roland Lauster
- Technische Universität Berlin, Medical Biotechnology, TIB 4/4-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Uwe Marx
- Technische Universität Berlin, Medical Biotechnology, TIB 4/4-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany; TissUse GmbH, Markgrafenstraße 18, 15528 Spreenhagen, Germany.
| |
Collapse
|
86
|
Mechanisms of Vessel Pruning and Regression. Dev Cell 2015; 34:5-17. [PMID: 26151903 DOI: 10.1016/j.devcel.2015.06.004] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/26/2015] [Accepted: 06/03/2015] [Indexed: 01/27/2023]
Abstract
The field of angiogenesis research has primarily focused on the mechanisms of sprouting angiogenesis. Yet vascular networks formed by vessel sprouting subsequently undergo extensive vascular remodeling to form a functional and mature vasculature. This "trimming" includes distinct processes of vascular pruning, the regression of selected vascular branches. In some situations complete vascular networks may undergo physiological regression. Vessel regression is an understudied yet emerging field of research. This review summarizes the state-of-the-art of vessel pruning and regression with a focus on the cellular processes and the molecular regulators of vessel maintenance and regression.
Collapse
|
87
|
Hung OY, Brown AJ, Ahn SG, Veneziani A, Giddens DP, Samady H. Association of Wall Shear Stress with Coronary Plaque Progression and Transformation. Interv Cardiol Clin 2015; 4:491-502. [PMID: 28581935 DOI: 10.1016/j.iccl.2015.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coronary endothelial function regulation by wall shear stress (WSS), the frictional force of blood exerted against the vessel wall, can help explain the focal propensity of plaque development in an environment of systemic atherosclerosis risk factors. Sustained abnormal pathologic WSS leads to a proatherogenic endothelial cell phenotype, plaque progression and transformation, and adaptive vascular remodeling in site-specific areas. Assessing dynamic coronary plaque compositional changes in vivo remains challenging; however, recent advances in intravascular image acquisition and processing may provide swifter WSS calculations and make possible larger prospective investigations on the prognostic value of WSS in patients with coronary atherosclerosis.
Collapse
Affiliation(s)
- Olivia Y Hung
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road NE, Atlanta, GA 30322, USA
| | - Adam J Brown
- Department of Cardiovascular Medicine, University of Cambridge, ACCI Level 6, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Sung Gyun Ahn
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road NE, Atlanta, GA 30322, USA; Division of Cardiology, Yonsei University, Wonju College of Medicine, 20 Ilsan-ro, Wonju 220-701, South Korea
| | - Alessandro Veneziani
- Department of Mathematics and Computer Science, Emory University, 400 Dowman Drive, Atlanta, GA 30322, USA
| | - Don P Giddens
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr, Atlanta, GA 30332, USA
| | - Habib Samady
- Interventional Cardiology, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road, Suite F606, Atlanta, GA 30322, USA.
| |
Collapse
|
88
|
Novel insights into the regulation of cyclooxygenase-2 expression by platelet-cancer cell cross-talk. Biochem Soc Trans 2015; 43:707-14. [PMID: 26551717 PMCID: PMC4613509 DOI: 10.1042/bst20140322] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Indexed: 12/16/2022]
Abstract
Platelets are activated by the interaction with cancer cells and release enhanced levels of lipid mediators [such as thromboxane (TX)A2 and prostaglandin (PG)E2, generated from arachidonic acid (AA) by the activity of cyclooxygenase (COX)-1], granule content, including ADP and growth factors, chemokines, proteases and Wnt proteins. Moreover, activated platelets shed different vesicles, such as microparticles (MPs) and exosomes (rich in genetic material such as mRNAs and miRNAs). These platelet-derived products induce several phenotypic changes in cancer cells which confer high metastatic capacity. A central event involves an aberrant expression of COX-2 which influences cell-cycle progression and contribute to the acquisition of a cell migratory phenotype through the induction of epithelial mesenchymal transition genes and down-regulation of E-cadherin expression. The identification of novel molecular determinants involved in the cross-talk between platelets and cancer cells has led to identify novel targets for anti-cancer drug development.
Collapse
|
89
|
Kanthi Y, Hyman MC, Liao H, Baek AE, Visovatti SH, Sutton NR, Goonewardena SN, Neral MK, Jo H, Pinsky DJ. Flow-dependent expression of ectonucleotide tri(di)phosphohydrolase-1 and suppression of atherosclerosis. J Clin Invest 2015; 125:3027-36. [PMID: 26121751 DOI: 10.1172/jci79514] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/21/2015] [Indexed: 01/18/2023] Open
Abstract
The ability of cells to detect and respond to nucleotide signals in the local microenvironment is essential for vascular homeostasis. The enzyme ectonucleotide tri(di)phosphohydrolase-1 (ENTPD1, also known as CD39) on the surface of leukocytes and endothelial cells metabolizes locally released, intravascular ATP and ADP, thereby eliminating these prothrombotic and proinflammatory stimuli. Here, we evaluated the contribution of CD39 to atherogenesis in the apolipoprotein E-deficient (ApoE-deficient) mouse model of atherosclerosis. Compared with control ApoE-deficient animals, plaque burden was markedly increased along with circulating markers of platelet activation in Cd39+/-Apoe-/- mice fed a high-fat diet. Plaque analysis revealed stark regionalization of endothelial CD39 expression and function in Apoe-/- mice, with CD39 prominently expressed in atheroprotective, stable flow regions and diminished in atheroprone areas subject to disturbed flow. In mice, disturbed flow as the result of partial carotid artery ligation rapidly suppressed endothelial CD39 expression. Moreover, unidirectional laminar shear stress induced atheroprotective CD39 expression in human endothelial cells. CD39 induction was dependent upon the vascular transcription factor Krüppel-like factor 2 (KLF2) binding near the transcriptional start site of CD39. Together, these data establish CD39 as a regionalized regulator of atherogenesis that is driven by shear stress.
Collapse
|
90
|
Filipovic N, Ghimire K, Saveljic I, Milosevic Z, Ruegg C. Computational modeling of shear forces and experimental validation of endothelial cell responses in an orbital well shaker system. Comput Methods Biomech Biomed Engin 2015; 19:581-90. [DOI: 10.1080/10255842.2015.1051973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
91
|
Dick M, MacDonald K, Tardif JC, Leask RL. The effect of simvastatin treatment on endothelial cell response to shear stress and tumor necrosis factor alpha stimulation. Biomed Eng Online 2015; 14:58. [PMID: 26091905 PMCID: PMC4475335 DOI: 10.1186/s12938-015-0057-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/12/2015] [Indexed: 11/15/2022] Open
Abstract
Background Statin drugs are one of the most commonly prescribed pharmaceuticals by physicians. By blocking the rate-limiting step in the cholesterol biosynthesis pathway, statins inhibit cholesterol synthesis, which benefits patient health. However, since many other important cellular processes are regulated within this pathway, they may also be influenced by statin therapy. These pleiotropic effects of statins have not been fully investigated, but are believed to positively influence endothelial cells (ECs), which line the vasculature in a confluent monolayer. Few studies have considered the effect of blood flow on ECs and how this may augment EC response to statins. Methods In this study, the effect of statin treatment on ECs is investigated for cells stimulated with tumor necrosis factor alpha (TNF-α), an inflammatory cytokine that promotes an atheroprone endothelium. Additionally, ECs are exposed to a physiologically relevant wall shear stress (WSS) of 12.5 dynes/cm2 using a three-dimensional tissue culture model to provide a realistic hemodynamic environment. ECs are analyzed for morphology using light microscopy as well as cytoskeletal structure and alignment using confocal microscopy. Statistical analysis is performed on the results using both the one-way analysis of variance with Bonferroni post-tests and the two-tailed t test. Results We have shown that statin treatment caused cells to adapt to a rounded, atheroprone morphology, with a significantly higher shape index. Oppositely, TNF-α stimulation caused cells to elongate to an atheroprotective morphology, with a significantly lower shape index. WSS and TNF-α were unable to reverse any statin-induced cell rounding or F-actin disruption. Conclusion Further work is therefore needed to determine why statin drugs cause cells to have an atheroprone morphology, but an atheroprotective genotype, and why TNF-α stimulation causes an atheroprotective morphology, but an atheroprone genotype. Despite the morphological changes due to statins or stimulation, ECs still respond to WSS. Understanding how statins influence ECs will allow for more targeted treatments for hypercholestemia and potentially other diseases.
Collapse
Affiliation(s)
- Melissa Dick
- Department of Chemical Engineering, McGill University, 3610 University Avenue, Montreal, QC, H3A 2B2, Canada. .,Montreal Heart Institute, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.
| | - Katherine MacDonald
- Department of Chemical Engineering, McGill University, 3610 University Avenue, Montreal, QC, H3A 2B2, Canada.
| | - Jean-Claude Tardif
- Montreal Heart Institute, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.
| | - Richard L Leask
- Department of Chemical Engineering, McGill University, 3610 University Avenue, Montreal, QC, H3A 2B2, Canada. .,Montreal Heart Institute, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.
| |
Collapse
|
92
|
Sacerdoti D, Pesce P, Di Pascoli M, Brocco S, Cecchetto L, Bolognesi M. Arachidonic acid metabolites and endothelial dysfunction of portal hypertension. Prostaglandins Other Lipid Mediat 2015; 120:80-90. [PMID: 26072731 DOI: 10.1016/j.prostaglandins.2015.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/20/2015] [Accepted: 05/25/2015] [Indexed: 12/12/2022]
Abstract
Increased resistance to portal flow and increased portal inflow due to mesenteric vasodilatation represent the main factors causing portal hypertension in cirrhosis. Endothelial cell dysfunction, defined as an imbalance between the synthesis, release, and effect of endothelial mediators of vascular tone, inflammation, thrombosis, and angiogenesis, plays a major role in the increase of resistance in portal circulation, in the decrease in the mesenteric one, in the development of collateral circulation. Reduced response to vasodilators in liver sinusoids and increased response in the mesenteric arterioles, and, viceversa, increased response to vasoconstrictors in the portal-sinusoidal circulation and decreased response in the mesenteric arterioles are also relevant to the pathophysiology of portal hypertension. Arachidonic acid (AA) metabolites through the three pathways, cyclooxygenase (COX), lipoxygenase, and cytochrome P450 monooxygenase and epoxygenase, are involved in endothelial dysfunction of portal hypertension. Increased thromboxane-A2 production by liver sinusoidal endothelial cells (LSECs) via increased COX-1 activity/expression, increased leukotriens, increased epoxyeicosatrienoic acids (EETs) (dilators of the peripheral arterial circulation, but vasoconstrictors of the portal-sinusoidal circulation), represent a major component in the increased portal resistance, in the decreased portal response to vasodilators and in the hyper-response to vasoconstrictors. Increased prostacyclin (PGI2) via COX-1 and COX-2 overexpression, and increased EETs/heme-oxygenase-1/K channels/gap junctions (endothelial derived hyperpolarizing factor system) play a major role in mesenteric vasodilatation, hyporeactivity to vasoconstrictors, and hyper-response to vasodilators. EETs, mediators of liver regeneration after hepatectomy and of angiogenesis, may play a role in the development of regenerative nodules and collateral circulation, through stimulation of vascular endothelial growth factor (VEGF) inside the liver and in the portal circulation. Pharmacological manipulation of AA metabolites may be beneficial for cirrhotic portal hypertension.
Collapse
Affiliation(s)
- David Sacerdoti
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy.
| | - Paola Pesce
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Marco Di Pascoli
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Silvia Brocco
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Lara Cecchetto
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Massimo Bolognesi
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| |
Collapse
|
93
|
|
94
|
Miano JM, Long X. The short and long of noncoding sequences in the control of vascular cell phenotypes. Cell Mol Life Sci 2015; 72:3457-88. [PMID: 26022065 DOI: 10.1007/s00018-015-1936-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022]
Abstract
The two principal cell types of importance for normal vessel wall physiology are smooth muscle cells and endothelial cells. Much progress has been made over the past 20 years in the discovery and function of transcription factors that coordinate proper differentiation of these cells and the maintenance of vascular homeostasis. More recently, the converging fields of bioinformatics, genomics, and next generation sequencing have accelerated discoveries in a number of classes of noncoding sequences, including transcription factor binding sites (TFBS), microRNA genes, and long noncoding RNA genes, each of which mediates vascular cell differentiation through a variety of mechanisms. Alterations in the nucleotide sequence of key TFBS or deviations in transcription of noncoding RNA genes likely have adverse effects on normal vascular cell phenotype and function. Here, the subject of noncoding sequences that influence smooth muscle cell or endothelial cell phenotype will be summarized as will future directions to further advance our understanding of the increasingly complex molecular circuitry governing normal vascular cell differentiation and how such information might be harnessed to combat vascular diseases.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA,
| | | |
Collapse
|
95
|
Dunn J, Simmons R, Thabet S, Jo H. The role of epigenetics in the endothelial cell shear stress response and atherosclerosis. Int J Biochem Cell Biol 2015; 67:167-76. [PMID: 25979369 DOI: 10.1016/j.biocel.2015.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 04/30/2015] [Accepted: 05/02/2015] [Indexed: 12/15/2022]
Abstract
Currently in the field of vascular biology, the role of epigenetics in endothelial cell biology and vascular disease has attracted more in-depth study. Using both in vitro and in vivo models of blood flow, investigators have recently begun to reveal the underlying epigenetic regulation of endothelial gene expression. Recently, our group, along with two other independent groups, have demonstrated that blood flow controls endothelial gene expression by DNA methyltransferases (DNMT1 and 3A). Disturbed flow (d-flow), characterized by low and oscillating shear stress (OS), is pro-atherogenic and induces expression of DNMT1 both in vivo and in vitro. D-flow regulates genome-wide DNA methylation patterns in a DNMT-dependent manner. The DNMT inhibitor 5-Aza-2'deoxycytidine (5Aza) or DNMT1 siRNA reduces OS-induced endothelial inflammation. Moreover, 5Aza inhibits the development of atherosclerosis in ApoE(-/-) mice. Through a systems biological analysis of genome-wide DNA methylation patterns and gene expression data, we found 11 mechanosensitive genes which were suppressed by d-flow in vivo, experienced hypermethylation in their promoter region in response to d-flow, and were rescued by 5Aza treatment. Interestingly, among these mechanosensitive genes, the two transcription factors HoxA5 and Klf3 contain cAMP-response-elements (CRE), which may indicate that methylation of CRE sites could serve as a mechanosensitive master switch in gene expression. These findings provide new insight into the mechanism by which flow controls epigenetic DNA methylation patterns, which in turn alters endothelial gene expression, regulates vascular biology, and induces atherosclerosis. These novel findings have broad implications for understanding the biochemical mechanisms of atherogenesis and provide a basis for identifying potential therapeutic targets for atherosclerosis. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Jessilyn Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Rachel Simmons
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Salim Thabet
- Division of Cardiology, Georgia Institute of Technology and Emory University, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA; Division of Cardiology, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
96
|
Leyendecker G, Wildt L. A new concept of endometriosis and adenomyosis: tissue injury and repair (TIAR). Horm Mol Biol Clin Investig 2015; 5:125-42. [PMID: 25961248 DOI: 10.1515/hmbci.2011.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 01/12/2011] [Indexed: 01/10/2023]
Abstract
Pelvic endometriosis, deeply infiltrating endometriosis and uterine adenomyosis share a common pathophysiology and may be integrated into the physiological mechanism and new nosological concept of 'tissue injury and repair' (TIAR) and may, in this context, just represent the extreme of a basically physiological, estrogen-related mechanism that is pathologically exaggerated in an extremely estrogen-sensitive reproductive organ. The acronym TIAR describes a fundamental and apparently ubiquitous biological system that becomes operative in mesenchymal tissues following tissue injury and, upon activation, results in the local production of estradiol. Endometriosis and adenomyosis are caused by trauma. In the spontaneously developing disease, chronic uterine peristaltic activity or phases of hyperperistalsis induce, at the endometrial-myometrial interface near the fundo-cornual raphe, microtraumatisations, with activation of the TIAR mechanism. With ongoing traumatisations, such sites of inflammation might accumulate and the increasingly produced estrogens interfere in a paracrine fashion with ovarian control over uterine peristaltic activity, resulting in permanent hyperperistalsis and a self-perpetuation of the disease process. Overt autotraumatisation of the uterus with dislocation of fragments of basal endometrium into the peritoneal cavity and infiltration of basal endometrium into the depth of the myometrial wall ensues. In most cases of endometriosis/adenomyosis a causal event early in the reproductive period of life must be postulated, rapidly leading to archimetral hyperestrogenism and uterine hyperperistalsis. In late premenopausal adenomyosis such an event might not have occurred. However, as indicated by the high prevalence of the disease, it appears to be unavoidable that, with time, chronic normoperistalsis throughout the reproductive period of life accumulates to the same extent of microtraumatisation. With activation of the TIAR mechanism followed by chronic inflammation and infiltrative growth, endometriosis/adenomyosis of the younger woman and premenopausal adenomyosis share in principal the same pathophysiology.
Collapse
|
97
|
Dunn J, Thabet S, Jo H. Flow-Dependent Epigenetic DNA Methylation in Endothelial Gene Expression and Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1562-9. [PMID: 25953647 DOI: 10.1161/atvbaha.115.305042] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/27/2015] [Indexed: 12/31/2022]
Abstract
Epigenetic mechanisms that regulate endothelial cell gene expression are now emerging. DNA methylation is the most stable epigenetic mark that confers persisting changes in gene expression. Not only is DNA methylation important in rendering cell identity by regulating cell type-specific gene expression throughout differentiation, but it is becoming clear that DNA methylation also plays a key role in maintaining endothelial cell homeostasis and in vascular disease development. Disturbed blood flow causes atherosclerosis, whereas stable flow protects against it by differentially regulating gene expression in endothelial cells. Recently, we and others have shown that flow-dependent gene expression and atherosclerosis development are regulated by mechanisms dependent on DNA methyltransferases (1 and 3A). Disturbed blood flow upregulates DNA methyltransferase expression both in vitro and in vivo, which leads to genome-wide DNA methylation alterations and global gene expression changes in a DNA methyltransferase-dependent manner. These studies revealed several mechanosensitive genes, such as HoxA5, Klf3, and Klf4, whose promoters were hypermethylated by disturbed blood flow, but rescued by DNA methyltransferases inhibitors such as 5Aza-2-deoxycytidine. These findings provide new insight into the mechanism by which flow controls epigenomic DNA methylation patterns, which in turn alters endothelial gene expression, regulates vascular biology, and modulates atherosclerosis development.
Collapse
Affiliation(s)
- Jessilyn Dunn
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta
| | - Salim Thabet
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta
| | - Hanjoong Jo
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta.
| |
Collapse
|
98
|
Chu LY, Liou JY, Wu KK. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat 2015; 118-119:19-27. [PMID: 25910681 DOI: 10.1016/j.prostaglandins.2015.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/25/2015] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Vascular integrity is protected by the lining endothelial cells (ECs) through structural and molecular protective mechanisms. In response to external stresses, ECs are dynamic in producing protective molecules such as prostacyclin (PGI2). PGI2 is known to inhibit platelet aggregation and controls smooth muscle cell contraction via IP receptors. Recent studies indicate that PGI2 defends endothelial survival and protects vascular smooth muscle cell from apoptosis via peroxisome-proliferator activated receptors (PPAR). PPAR activation results in 14-3-3 upregulation. Increase in cytosolic 14-3-3ɛ or 14-3-3β enhances binding and sequestration of Akt-mediated phosphorylated Bad and reduces Bad-mediated apoptosis via the mitochondrial pathway. Experimental data indicate that administration of PGI2 analogs or augmentation of PGI2 production by gene transfer attenuates endothelial damage and organ infarction caused by ischemia-reperfusion injury. The protective effect of PGI2 is attributed in part to preserving endothelial integrity.
Collapse
Affiliation(s)
- Ling-yun Chu
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jun-Yang Liou
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan; Institute of Cell and System Medicine, National Health Research Institute, Chunan, Taiwan
| | - Kenneth K Wu
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan; Institute of Cell and System Medicine, National Health Research Institute, Chunan, Taiwan; Department of Medical Sciences, National Tsing-Hua University, Hsin-chu, Taiwan.
| |
Collapse
|
99
|
Diaz MF, Li N, Lee HJ, Adamo L, Evans SM, Willey HE, Arora N, Torisawa YS, Vickers DA, Morris SA, Naveiras O, Murthy SK, Ingber DE, Daley GQ, García-Cardeña G, Wenzel PL. Biomechanical forces promote blood development through prostaglandin E2 and the cAMP-PKA signaling axis. ACTA ACUST UNITED AC 2015; 212:665-80. [PMID: 25870199 PMCID: PMC4419354 DOI: 10.1084/jem.20142235] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/17/2015] [Indexed: 12/12/2022]
Abstract
Blood flow promotes emergence of definitive hematopoietic stem cells (HSCs) in the developing embryo, yet the signals generated by hemodynamic forces that influence hematopoietic potential remain poorly defined. Here we show that fluid shear stress endows long-term multilineage engraftment potential upon early hematopoietic tissues at embryonic day 9.5, an embryonic stage not previously described to harbor HSCs. Effects on hematopoiesis are mediated in part by a cascade downstream of wall shear stress that involves calcium efflux and stimulation of the prostaglandin E2 (PGE2)-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling axis. Blockade of the PGE2-cAMP-PKA pathway in the aorta-gonad-mesonephros (AGM) abolished enhancement in hematopoietic activity. Furthermore, Ncx1 heartbeat mutants, as well as static cultures of AGM, exhibit lower levels of expression of prostaglandin synthases and reduced phosphorylation of the cAMP response element-binding protein (CREB). Similar to flow-exposed cultures, transient treatment of AGM with the synthetic analogue 16,16-dimethyl-PGE2 stimulates more robust engraftment of adult recipients and greater lymphoid reconstitution. These data provide one mechanism by which biomechanical forces induced by blood flow modulate hematopoietic potential.
Collapse
Affiliation(s)
- Miguel F Diaz
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Nan Li
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Hyun Jung Lee
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Luigi Adamo
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Siobahn M Evans
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Hannah E Willey
- Department of Bioengineering, Rice University, Houston, TX 77030
| | - Natasha Arora
- Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Yu-Suke Torisawa
- Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 Wyss Institute for Biologically Inspired Engineering at Harvard University and Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115 Wyss Institute for Biologically Inspired Engineering at Harvard University and Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Dwayne A Vickers
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA 02115
| | - Samantha A Morris
- Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Olaia Naveiras
- Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Shashi K Murthy
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA 02115
| | - Donald E Ingber
- Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 Wyss Institute for Biologically Inspired Engineering at Harvard University and Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115 Wyss Institute for Biologically Inspired Engineering at Harvard University and Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - George Q Daley
- Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Children's Hospital Boston and Dana-Farber Cancer Institute, Boston, MA 02115 Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 Harvard Stem Cell Institute and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Pamela L Wenzel
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030 Program in Children's Regenerative Medicine, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, and Immunology Program, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
100
|
Davis CA, Zambrano S, Anumolu P, Allen ACB, Sonoqui L, Moreno MR. Device-Based In Vitro Techniques for Mechanical Stimulation of Vascular Cells: A Review. J Biomech Eng 2015; 137:040801. [DOI: 10.1115/1.4029016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/07/2014] [Indexed: 01/19/2023]
Abstract
The most common cause of death in the developed world is cardiovascular disease. For decades, this has provided a powerful motivation to study the effects of mechanical forces on vascular cells in a controlled setting, since these cells have been implicated in the development of disease. Early efforts in the 1970 s included the first use of a parallel-plate flow system to apply shear stress to endothelial cells (ECs) and the development of uniaxial substrate stretching techniques (Krueger et al., 1971, “An in Vitro Study of Flow Response by Cells,” J. Biomech., 4(1), pp. 31–36 and Meikle et al., 1979, “Rabbit Cranial Sutures in Vitro: A New Experimental Model for Studying the Response of Fibrous Joints to Mechanical Stress,” Calcif. Tissue Int., 28(2), pp. 13–144). Since then, a multitude of in vitro devices have been designed and developed for mechanical stimulation of vascular cells and tissues in an effort to better understand their response to in vivo physiologic mechanical conditions. This article reviews the functional attributes of mechanical bioreactors developed in the 21st century, including their major advantages and disadvantages. Each of these systems has been categorized in terms of their primary loading modality: fluid shear stress (FSS), substrate distention, combined distention and fluid shear, or other applied forces. The goal of this article is to provide researchers with a survey of useful methodologies that can be adapted to studies in this area, and to clarify future possibilities for improved research methods.
Collapse
Affiliation(s)
- Caleb A. Davis
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Steve Zambrano
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Pratima Anumolu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Alicia C. B. Allen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712-1801 e-mail:
| | - Leonardo Sonoqui
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Michael R. Moreno
- Department of Mechanical Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3123 e-mail:
| |
Collapse
|