1
|
Singh J, Ruhoff AM, Ashok D, Wise SG, Waterhouse A. Engineering advanced in vitro models of endothelial dysfunction. Trends Biotechnol 2025:S0167-7799(25)00089-7. [PMID: 40187930 DOI: 10.1016/j.tibtech.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Endothelial dysfunction is an important initiator of cardiovascular disease, the leading cause of death globally, and often manifests in arterial regions with disturbed blood flow. Experimental model advances have crucially helped unravel physiological mechanisms. While in vivo models provide a dynamic environment, they often fail to mimic human physiology precisely and face significant ethical barriers. Advanced in vitro models, including organs-on-chips and bioreactors, combine human cells and blood flow to accurately replicate endothelial dysfunction. Newer models have enhanced scalability and accuracy, with organs-on-chips commonly outperforming standard preclinical methods. Importantly, recent endothelial dysfunction discoveries leverage dynamic models to identify and evaluate clinically promising therapeutics. Here, we examine these developments and explore opportunities to develop next-generation in vitro models of endothelial dysfunction.
Collapse
Affiliation(s)
- Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Ruhoff
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Deepu Ashok
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Steven G Wise
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
2
|
Santos F, Sum H, Yan DCL, Brewer AC. Metaboloepigenetics: Role in the Regulation of Flow-Mediated Endothelial (Dys)Function and Atherosclerosis. Cells 2025; 14:378. [PMID: 40072106 PMCID: PMC11898952 DOI: 10.3390/cells14050378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Endothelial dysfunction is the main initiating factor in atherosclerosis. Through mechanotransduction, shear stress regulates endothelial cell function in both homeostatic and diseased states. Accumulating evidence reveals that epigenetic changes play critical roles in the etiology of cardiovascular diseases, including atherosclerosis. The metabolic regulation of epigenetics has emerged as an important factor in the control of gene expression in diseased states, but to the best of our knowledge, this connection remains largely unexplored in endothelial dysfunction and atherosclerosis. In this review, we (1) summarize how shear stress (or flow) regulates endothelial (dys)function; (2) explore the epigenetic alterations that occur in the endothelium in response to disturbed flow; (3) review endothelial cell metabolism under different shear stress conditions; and (4) suggest mechanisms which may link this altered metabolism to the regulation of the endothelial epigenome by modulations in metabolite availability. We believe that metabolic regulation plays an important role in endothelial epigenetic reprogramming and could pave the way for novel metabolism-based therapeutic strategies.
Collapse
Affiliation(s)
- Francisco Santos
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | - Hashum Sum
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | | | - Alison C. Brewer
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| |
Collapse
|
3
|
Blazeski A, Floryan MA, Zhang Y, Fajardo Ramírez OR, Meibalan E, Ortiz-Urbina J, Angelidakis E, Shelton SE, Kamm RD, García-Cardeña G. Engineering microvascular networks using a KLF2 reporter to probe flow-dependent endothelial cell function. Biomaterials 2024; 311:122686. [PMID: 38971122 DOI: 10.1016/j.biomaterials.2024.122686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/12/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Shear stress generated by the flow of blood in the vasculature is a potent regulator of endothelial cell function and vascular structure. While vascular responses to flow are complex and context-dependent, endothelial cell signaling in response to shear stress induced by laminar flows is coordinated by the transcription factor KLF2. The flow-dependent expression of KLF2 in endothelial cells is associated with a quiescent, anti-inflammatory phenotype and has been well characterized in two-dimensional systems but has not been studied in three-dimensional in vitro systems. Here we develop engineered microvascular networks (MVNs) that incorporate a KLF2-based endothelial cell flow sensor within a microfluidic chip, apply continuous flow using an attached microfluidic pump, and study the effects of this flow on vascular structure and function. We found that application of flow to MVNs for 48 h resulted in increased expression of the KLF2 reporter, larger vessel diameters, and decreased vascular branching and resistance. Notably, vessel diameters after the application of flow were independent of initial MVN morphologies. Finally, we found that MVNs exposed to flow have improved vascular barrier function and decreased platelet adhesion. MVNs with KLF2-based flow sensors represent a novel, powerful tool for evaluating the structural and functional effects of flow on engineered three-dimensional vascular systems.
Collapse
Affiliation(s)
- Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yuzhi Zhang
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA
| | - Oscar R Fajardo Ramírez
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA
| | - Elamaran Meibalan
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA
| | - Jesús Ortiz-Urbina
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA
| | - Emmanouil Angelidakis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, USA and Harvard Medical School, Boston, MA, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
El-Ansary A, Alhakbany M, Alfawaz HA, Al-Ayadhi LY. Indian Hedgehog (IHh) Protein and COX-2 as Biomarkers to Define the Mechanism of Epilepsy and Gastrointestinal Problems as Comorbid Medical Illnesses in Autism Spectrum Disorder: Combining ROC Curves to Improve Diagnostic Values. J Clin Med 2024; 13:6695. [PMID: 39597837 PMCID: PMC11595056 DOI: 10.3390/jcm13226695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Autism spectrum disorder (ASD) is a neurodevelopmental disorder that is increasing throughout the world. Nevertheless, no specific diagnostic or even risk assessment marker is available. Combining more than one marker can improve the diagnostic value of each marker alone and maximize the AUC for ROC curve. Hedgehog (Hh) signaling modulates both intestinal inflammation and immunity. Cyclooxygenase-2 (COX-2) is required for inflammation, and it has been related to epileptic seizures. COX-2 generates prostaglandins-EP2 (PGE2) in the brain, which plays a major role in neuropathology and epilepsy. This study aims to understand the pathophysiology of ASD by investigating the diagnostic value of COX-2 and IHh using independent and combined ROC curves. Methods: COX-2 and IHh were measured in 40 children with ASD and 41 age and sex-matched controls using ELISA. Statistical analyses were performed using ROC curves, odds ratios, and multiple logistic regression models. Results: Higher levels of COX-2 and IHh were observed in the plasma of patients with autism than in controls. The ROC curve of HIh and COX-2 independently showed poor and fair AUCs of 0.662 and 0.776, respectively, while the combined ROC for both variables in relation to the control group increased the AUC = 0.831 significantly (p < 0.001). Conclusions: Thus, combining these variables could be a useful diagnostic marker for ASD.
Collapse
Affiliation(s)
- Afaf El-Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, P.O. Box 110281, Abu Dhabi 23251, United Arab Emirates
| | - Manan Alhakbany
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11495, Saudi Arabia; (M.A.); (L.Y.A.-A.)
| | - Hanan A. Alfawaz
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| | - Laila Y. Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11495, Saudi Arabia; (M.A.); (L.Y.A.-A.)
| |
Collapse
|
5
|
Hu Y, Li W, Cheng X, Yang H, She ZG, Cai J, Li H, Zhang XJ. Emerging Roles and Therapeutic Applications of Arachidonic Acid Pathways in Cardiometabolic Diseases. Circ Res 2024; 135:222-260. [PMID: 38900855 DOI: 10.1161/circresaha.124.324383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Cardiometabolic disease has become a major health burden worldwide, with sharply increasing prevalence but highly limited therapeutic interventions. Emerging evidence has revealed that arachidonic acid derivatives and pathway factors link metabolic disorders to cardiovascular risks and intimately participate in the progression and severity of cardiometabolic diseases. In this review, we systemically summarized and updated the biological functions of arachidonic acid pathways in cardiometabolic diseases, mainly focusing on heart failure, hypertension, atherosclerosis, nonalcoholic fatty liver disease, obesity, and diabetes. We further discussed the cellular and molecular mechanisms of arachidonic acid pathway-mediated regulation of cardiometabolic diseases and highlighted the emerging clinical advances to improve these pathological conditions by targeting arachidonic acid metabolites and pathway factors.
Collapse
Affiliation(s)
- Yufeng Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Xu Cheng
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Hailong Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Zhi-Gang She
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Jingjing Cai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China (H.L.)
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- School of Basic Medical Sciences, Wuhan University, China (X.-J.Z.)
| |
Collapse
|
6
|
Macvanin MT, Gluvic ZM, Klisic AN, Manojlovic MS, Suri JS, Rizzo M, Isenovic ER. The Link between miRNAs and PCKS9 in Atherosclerosis. Curr Med Chem 2024; 31:6926-6956. [PMID: 37990898 DOI: 10.2174/0109298673262124231102042914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 11/23/2023]
Abstract
Cardiovascular disease (CDV) represents the major cause of death globally. Atherosclerosis, as the primary cause of CVD, is a chronic immune-inflammatory disorder with complex multifactorial pathophysiology encompassing oxidative stress, enhanced immune-inflammatory cascade, endothelial dysfunction, and thrombosis. An initiating event in atherosclerosis is the subendothelial accumulation of low-density lipoprotein (LDL), followed by the localization of macrophages to fatty deposits on blood vessel walls, forming lipid-laden macrophages (foam cells) that secrete compounds involved in plaque formation. Given the fact that foam cells are one of the key culprits that underlie the pathophysiology of atherosclerosis, special attention has been paid to the investigation of the efficient therapeutic approach to overcome the dysregulation of metabolism of cholesterol in macrophages, decrease the foam cell formation and/or to force its degradation. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secretory serine proteinase that has emerged as a significant regulator of the lipid metabolism pathway. PCSK9 activation leads to the degradation of LDL receptors (LDLRs), increasing LDL cholesterol (LDL-C) levels in the circulation. PCSK9 pathway dysregulation has been identified as one of the mechanisms involved in atherosclerosis. In addition, microRNAs (miRNAs) are investigated as important epigenetic factors in the pathophysiology of atherosclerosis and dysregulation of lipid metabolism. This review article summarizes the recent findings connecting the role of PCSK9 in atherosclerosis and the involvement of various miRNAs in regulating the expression of PCSK9-related genes. We also discuss PCSK9 pathway-targeting therapeutic interventions based on PCSK9 inhibition, and miRNA levels manipulation by therapeutic agents.
Collapse
Affiliation(s)
- Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran M Gluvic
- Department of Endocrinology and Diabetes, School of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, Clinic of Internal Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra N Klisic
- Faculty of Medicine, Center for Laboratory Diagnostic, Primary Health Care Center, University of Montenegro, Podgorica, Montenegro
| | - Mia S Manojlovic
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, Athero- Point™, Roseville, CA95661, USA
| | - Manfredi Rizzo
- Department of Health Promotion, School of Medicine, Mother and Child Care and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
7
|
Dash S, Singh PA, Bajwa N, Choudhury A, Bisht P, Sharma R. Why Pharmacovigilance of Non-steroidal Anti-inflammatory Drugs is Important in India? Endocr Metab Immune Disord Drug Targets 2024; 24:731-748. [PMID: 37855282 DOI: 10.2174/0118715303247469230926092404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/15/2023] [Accepted: 08/18/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Non-steroidal Anti-Inflammatory Drugs (NSAIDs) are among the drugs that are most regularly administered to manage inflammation and pain. Over-the-Counter (OTC) NSAIDs are widely accessible, particularly in developing countries like India. This casual approach to using NSAIDs may operate as a magnet for NSAID-related adverse drug reactions (ADRs) among patients. OBJECTIVES As patients in India are less informed about the appropriate use of NSAIDs and consumption patttern, adverse drug reactions, and the importance of reporting ADRs, the current study's objective is to promote patient safety by using pharmacovigilance as a tool to educate patients. METHODS A targeted literature methodology was utilized to gather the data pertaining to NSAIDs, their ADRs and their pharmacovigilance. Different scientific databases, such as Science Direct, PubMed, Wiley Online Library, Springer, and Google Scholar, along with authentic textbooks, were explored as reference literature. RESULTS In general, NSAIDs consumption pattern depends upon the different age groups. Around 1.6 billion tablets of NSAIDs are consumed in India for ailments, such as headaches, arthritis, menstrual cramps, osteoarthritis, back pain, rheumatoid arthritis, gout, osteoporosis, tendinitis, cancer pain and chronic pain. Common ADRs of NSAIDs include nausea, vomiting, headache, gastritis, abdominal pain, and diarrhoea. Also, they can cause renal damage and cardiovascular problems if not consumed in a dose-dependent manner. However, Diclofenac and Ibuprofen have both been linked to depression and dementia. There have been reports of aplastic anaemia, agranulocytosis linked to phenylbutazone, Stevens-Johnson, and Lyell's syndrome linked to isoxicam and piroxicam, as well as the vulnerability of new-borns to Reye's syndrome after aspirin use. Lack of awareness, time constraints and unpredictability, poor training in ADRs identification, etc., are some of the reasons for the under-reporting of ADR of NSAIDs in India. CONCLUSION In order to rationally prescribe NSAIDs, it is essential to be aware of probable ADR's and establish prescription guidelines. Prescribers' behaviour can be changed toward excellent prescribing practices by conducting routine prescription assessments dealing with NSAIDs and providing feedback. In the near future, it will be critical to strengthen ADR data management and expand the reach of pharmacovigilance programs, ADR monitoring centers, and healthcare professionals' especially pharmacists' training in rural locations.
Collapse
Affiliation(s)
- Subhransu Dash
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali, 140413, Punjab, India
| | - Preet Amol Singh
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali, 140413, Punjab, India
| | - Neha Bajwa
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali, 140413, Punjab, India
| | - Abinash Choudhury
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali, 140413, Punjab, India
| | - Preeti Bisht
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali, 140413, Punjab, India
| | - Rajiv Sharma
- College of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
8
|
Blankfield RP. Is fluid retention a cardiovascular risk factor? Clin Hemorheol Microcirc 2024; 88:277-288. [PMID: 39302357 PMCID: PMC11492017 DOI: 10.3233/ch-242128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Endothelial dysfunction, the earliest manifestation of atherosclerosis, can be initiated by both biochemicals and biomechanical forces. Atherosclerosis occurs predominantly at arterial branch points, arterial bifurcations and the curved segments of great arteries. These are the regions that blood flows turbulently. Turbulence promotes endothelial dysfunction by reducing shear stress upon endothelial cells. The endothelial glycocalyx mediates the effect of shear stress upon the endothelium. A mathematical analysis of cardiovascular hemodynamics demonstrates that fluid retention increases turbulence of blood flow. While there is no empirical data confirming this relationship, fluid retention is associated with adverse cardiovascular events. Every medical condition that causes fluid retention is associated with increased risk of both atherosclerotic cardiovascular disease and venous thromboembolic disease. In addition, most medications that cause fluid retention are associated with increased adverse cardiovascular effects. Calcium channel blockers (CCBs) and pioglitazone are exceptions to this generalization. Even though data regarding CCBs and pioglitazone contradict the hypothesis that fluid retention is a cardiovascular risk factor, these medications have favorable cardiovascular properties which may outweigh the negative effect of fluid retention. Determining whether or not fluid retention is a cardiovascular risk factor would require empirical data demonstrating a relationship between fluid retention and turbulence of blood flow. While this issue should be relevant to cardiovascular researchers, clinicians and patients, it is especially pertinent to the pharmaceutical industry. Four-dimensional magnetic resonance imaging and vector flow Doppler ultrasound have the capability to quantify turbulence of blood flow. These technologies could be utilized to settle the matter.
Collapse
Affiliation(s)
- Robert P. Blankfield
- Department of Family Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
9
|
Rongchang F, Kun W, Hui W. Study of the mechanism of action of sand therapy on atherosclerosis based on the two-phase flow-Casson model. Biomed Mater Eng 2024; 35:165-178. [PMID: 38043001 DOI: 10.3233/bme-230134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
BACKGROUND Sand therapy is a non-pharmacological physiotherapy method that uses the natural environment and resources of Xinjiang to treat through the heat transfer and magnetic effects of sand. OBJECTIVE Employing the two-phase flow-Casson blood flow model, we investigate the mechanism of atherosclerosis prevention via sand therapy, offering a biomechanical theoretical rationale for the prevention of atherosclerosis through sand therapy via the prism of computational fluid dynamics (CFD). METHODS Sand therapy experiments were conducted to obtain popliteal artery blood flow velocity, and blood was considered as a two-phase flow composed of plasma and red blood cells, and CFD method was applied to analyze the hemodynamic effects of Casson's blood viscosity model before and after sand therapy. RESULTS (1) The blood flow velocity increased by 0.24 m/s and 0.04 m/s at peak systolic and diastolic phases, respectively, after sand therapy; the axial velocity of blood vessels increased by 28.56% after sand therapy. (2) The average red blood cell viscosity decreased by 0.00014 Pa ⋅ s after sand therapy. (3) The low wall shear stress increased by 1.09 Pa and the high wall shear stress reached 41.47 Pa after sand therapy. (4) The time-averaged wall shear stress, shear oscillation index and relative retention time were reduced after sand therapy. CONCLUSION The increase of blood flow velocity after sand therapy can reduce the excessive deposition of cholesterol and other substances, the decrease of erythrocyte viscosity is beneficial to the migration of erythrocytes to the vascular center, the increase of low wall shear stress has a positive effect on the prevention of atherosclerosis, and the decrease of time-averaged wall shear stress, shear oscillation index and relative retention time can reduce the occurrence of thrombosis.
Collapse
Affiliation(s)
- Fu Rongchang
- College of Intelligent Manufacturing Modern Industry, School of Mechanical Engineering, Xinjiang University, Urumqi, China
| | - Wang Kun
- College of Intelligent Manufacturing Modern Industry, School of Mechanical Engineering, Xinjiang University, Urumqi, China
| | - Wu Hui
- College of Intelligent Manufacturing Modern Industry, School of Mechanical Engineering, Xinjiang University, Urumqi, China
| |
Collapse
|
10
|
Vinokurova M, Lopes-Pires ME, Cypaite N, Shala F, Armstrong PC, Ahmetaj-Shala B, Elghazouli Y, Nüsing R, Liu B, Zhou Y, Hao CM, Herschman HR, Mitchell JA, Kirkby NS. Widening the Prostacyclin Paradigm: Tissue Fibroblasts Are a Critical Site of Production and Antithrombotic Protection. Arterioscler Thromb Vasc Biol 2024; 44:271-286. [PMID: 37823267 PMCID: PMC10749679 DOI: 10.1161/atvbaha.123.318923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Prostacyclin is a fundamental signaling pathway traditionally associated with the cardiovascular system and protection against thrombosis but which also has regulatory functions in fibrosis, proliferation, and immunity. Prevailing dogma states that prostacyclin is principally derived from vascular endothelium, although it is known that other cells can also synthesize it. However, the role of nonendothelial sources in prostacyclin production has not been systematically evaluated resulting in an underappreciation of their importance relative to better characterized endothelial sources. METHODS To address this, we have used novel endothelial cell-specific and fibroblast-specific COX (cyclo-oxygenase) and prostacyclin synthase knockout mice and cells freshly isolated from mouse and human lung tissue. We have assessed prostacyclin release by immunoassay and thrombosis in vivo using an FeCl3-induced carotid artery injury model. RESULTS We found that in arteries, endothelial cells are the main source of prostacyclin but that in the lung, and other tissues, prostacyclin production occurs largely independently of endothelial and vascular smooth muscle cells. Instead, in mouse and human lung, prostacyclin production was strongly associated with fibroblasts. By comparison, microvascular endothelial cells from the lung showed weak prostacyclin synthetic capacity compared with those isolated from large arteries. Prostacyclin derived from fibroblasts and other nonendothelial sources was seen to contribute to antithrombotic protection. CONCLUSIONS These observations define a new paradigm in prostacyclin biology in which fibroblast/nonendothelial-derived prostacyclin works in parallel with endothelium-derived prostanoids to control thrombotic risk and potentially a broad range of other biology. Although generation of prostacyclin by fibroblasts has been shown previously, the scale and systemic activity was unappreciated. As such, this represents a basic change in our understanding and may provide new insight into how diseases of the lung result in cardiovascular risk.
Collapse
Affiliation(s)
- Maria Vinokurova
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Maria Elisa Lopes-Pires
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Neringa Cypaite
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Fisnik Shala
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Paul C. Armstrong
- Blizard Institute, Queen Mary University of London, United Kingdom (P.C.A.)
| | - Blerina Ahmetaj-Shala
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Youssef Elghazouli
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Rolf Nüsing
- Clinical Pharmacology and Pharmacotherapy Department, Goethe University, Frankfurt, Germany (R.N.)
| | - Bin Liu
- Cardiovascular Research Centre, Shantou University Medical College, China (B.L., Y.Z.)
| | - Yingbi Zhou
- Cardiovascular Research Centre, Shantou University Medical College, China (B.L., Y.Z.)
| | - Chuan-ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China (C.-m.H.)
| | - Harvey R. Herschman
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (H.R.H.)
| | - Jane A. Mitchell
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| | - Nicholas S. Kirkby
- National Heart and Lung Institute, Imperial College London, United Kingdom (M.V., M.E.L.-P., N.C., F.S., B.A.-S., Y.E., J.A.M., N.S.K.)
| |
Collapse
|
11
|
Blazeski A, Floryan MA, Fajardo-Ramírez OR, Meibalan E, Ortiz-Urbina J, Angelidakis E, Shelton SE, Kamm RD, García-Cardeña G. Engineering microvascular networks using a KLF2 reporter to probe flow-dependent endothelial cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.565021. [PMID: 37961543 PMCID: PMC10635035 DOI: 10.1101/2023.10.31.565021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Shear stress generated by the flow of blood in the vasculature is a potent regulator of endothelial cell phenotype and vascular structure. While vascular responses to flow are complex and context-dependent, endothelial cell signaling in response to shear stress induced by laminar flows is coordinated by the transcription factor KLF2. The expression of KLF2 in endothelial cells is associated with a quiescent, anti-inflammatory phenotype and has been well characterized in two-dimensional systems, but has not been studied in three-dimensional in vitro systems. Here we develop engineered microvascular networks (MVNs) with a KLF2-based endothelial cell sensor within a microfluidic chip, apply continuous flow using an attached microfluidic pump, and study the effects of this flow on vascular structure and function. We found that culture of MVNs exposed to flow for 48 hours that resulted in increased expression of the KLF2-GFP-reporter display larger vessel diameters and decreased vascular branching and resistance. Additionally, vessel diameters after the application of flow were independent of initial MVN morphologies. Finally, we found that MVNs exposed to flow have improved vascular barrier function and decreased platelet adhesion. The MVNs with KLF2-based flow sensors represent a powerful tool for evaluating the structural and functional effects of flow on engineered three-dimensional vascular systems.
Collapse
Affiliation(s)
- Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A. Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Oscar R. Fajardo-Ramírez
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
| | - Elamaran Meibalan
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
| | - Jesús Ortiz-Urbina
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emmanouil Angelidakis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E. Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
12
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
13
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
14
|
Hamrangsekachaee M, Wen K, Yazdani N, Willits RK, Bencherif SA, Ebong EE. Endothelial glycocalyx sensitivity to chemical and mechanical sub-endothelial substrate properties. Front Bioeng Biotechnol 2023; 11:1250348. [PMID: 38026846 PMCID: PMC10643223 DOI: 10.3389/fbioe.2023.1250348] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Glycocalyx (GCX) is a carbohydrate-rich structure that coats the surface of endothelial cells (ECs) and lines the blood vessel lumen. Mechanical perturbations in the vascular environment, such as blood vessel stiffness, can be transduced and sent to ECs through mechanosensors such as GCX. Adverse stiffness alters GCX-mediated mechanotransduction and leads to EC dysfunction and eventually atherosclerotic cardiovascular diseases. To understand GCX-regulated mechanotransduction events, an in vitro model emulating in vivo vessel conditions is needed. To this end, we investigated the impact of matrix chemical and mechanical properties on GCX expression via fabricating a tunable non-swelling matrix based on the collagen-derived polypeptide, gelatin. To study the effect of matrix composition, we conducted a comparative analysis of GCX expression using different concentrations (60-25,000 μg/mL) of gelatin and gelatin methacrylate (GelMA) in comparison to fibronectin (60 μg/mL), a standard coating material for GCX-related studies. Using immunocytochemistry analysis, we showed for the first time that different substrate compositions and concentrations altered the overall GCX expression on human umbilical vein ECs (HUVECs). Subsequently, GelMA hydrogels were fabricated with stiffnesses of 2.5 and 5 kPa, representing healthy vessel tissues, and 10 kPa, corresponding to diseased vessel tissues. Immunocytochemistry analysis showed that on hydrogels with different levels of stiffness, the GCX expression in HUVECs remained unchanged, while its major polysaccharide components exhibited dysregulation in distinct patterns. For example, there was a significant decrease in heparan sulfate expression on pathological substrates (10 kPa), while sialic acid expression increased with increased matrix stiffness. This study suggests the specific mechanisms through which GCX may influence ECs in modulating barrier function, immune cell adhesion, and mechanotransduction function under distinct chemical and mechanical conditions of both healthy and diseased substrates.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
| | - Narges Yazdani
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Rebecca K. Willits
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Eno E. Ebong
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Neuroscience Department, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Costa-Filho H, Sales T, Paula S, Nicolau L, Queiroga M, Havt A, Soares P, Barbosa A, Souza M. Role of cyclooxygenases 1 and 2 in the maintenance of colonic mucosal integrity in an experimental colitis model. Braz J Med Biol Res 2023; 56:e12946. [PMID: 37909497 PMCID: PMC10609549 DOI: 10.1590/1414-431x2023e12946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/14/2023] [Indexed: 11/03/2023] Open
Abstract
The role of cyclooxygenase (COXs) isoforms in maintaining colonic mucosal integrity is not fully understood. This study aimed to evaluate the role of COX-1 and -2 on colonic mucosal integrity in an experimental colitis model. Colitis was induced in Wistar rats by intracolonic administration of 2,4,6-trinitrobenzenesulfonic acid (20 mg + 50% ethanol). The control group (sham group) received saline only. After 7, 14, or 28 days, colonic samples were removed, and macroscopic lesion scores, wet weight, myeloperoxidase activity, and transepithelial electrical resistance (TER) were determined. In other rat groups, colonic samples from the sham group and a 7th day post-colitis group were mounted in Üssing chambers with the luminal side exposed to a buffer solution (control), acetylsalicylic acid (ASA), SC-560 (COX-1 inhibitor), or celecoxib (COX-2 inhibitor). TER and epithelial permeability to fluorescein were measured. The 7th day colitis group had higher macroscopic damage scores, wet weight, and myeloperoxidase activity and lower basal TER than the sham, 14th day colitis, and 28th day colitis groups. Inhibition of COX-1 but not COX-2 significantly decreased TER and increased permeability to fluorescein in the 7th day post-colitis group compared to the sham group. Additionally, ASA decreased the colonic mucosal integrity on day seven post-colitis compared to the sham group. A decrease in the colonic mucosa integrity in the experimental colitis model can be aggravated only by the inhibition of COX-1, which demonstrated the importance of this enzyme in the maintenance of colonic mucosal integrity.
Collapse
Affiliation(s)
- H.B. Costa-Filho
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T.M.A.L. Sales
- Departamento de Medicina, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S.M. Paula
- Departamento de Medicina, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L.A.D. Nicolau
- Centro de Pesquisa em Biotecnologia e Biodiversidade, BIOTEC, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brasil
| | - M.L. Queiroga
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A. Havt
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P.M.G. Soares
- Departamento de Morfologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A.L.R. Barbosa
- Departamento de Fisioterapia, Universidade Federal do Delta do Parnaíba, Parnaíba, PI, Brasil
| | - M.H.L.P. Souza
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Medicina, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
16
|
Wang Y, Zhao ZG, Chai Z, Fang JC, Chen M. Electromagnetic field and cardiovascular diseases: A state-of-the-art review of diagnostic, therapeutic, and predictive values. FASEB J 2023; 37:e23142. [PMID: 37650634 DOI: 10.1096/fj.202300201rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
Despite encouraging advances in early diagnosis and treatment, cardiovascular diseases (CVDs) remained a leading cause of morbidity and mortality worldwide. Increasing evidence has shown that the electromagnetic field (EMF) influences many biological processes, which has attracted much attention for its potential therapeutic and diagnostic modalities in multiple diseases, such as musculoskeletal disorders and neurodegenerative diseases. Nonionizing EMF has been studied as a therapeutic or diagnostic tool in CVDs. In this review, we summarize the current literature ranging from in vitro to clinical studies focusing on the therapeutic potential (external EMF) and diagnostic potential (internal EMF generated from the heart) of EMF in CVDs. First, we provided an overview of the therapeutic potential of EMF and associated mechanisms in the context of CVDs, including cardiac arrhythmia, myocardial ischemia, atherosclerosis, and hypertension. Furthermore, we investigated the diagnostic and predictive value of magnetocardiography in CVDs. Finally, we discussed the critical steps necessary to translate this promising approach into clinical practice.
Collapse
Affiliation(s)
- Yan Wang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen-Gang Zhao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Chai
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian-Cheng Fang
- School of Instrumentation Science and Opto-Electronics Engineering, Beihang University, Beijing, China
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Ibrahim DM, Fomina A, Bouten CVC, Smits AIPM. Functional regeneration at the blood-biomaterial interface. Adv Drug Deliv Rev 2023; 201:115085. [PMID: 37690484 DOI: 10.1016/j.addr.2023.115085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/01/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The use of cardiovascular implants is commonplace in clinical practice. However, reproducing the key bioactive and adaptive properties of native cardiovascular tissues with an artificial replacement is highly challenging. Exciting new treatment strategies are under development to regenerate (parts of) cardiovascular tissues directly in situ using immunomodulatory biomaterials. Direct exposure to the bloodstream and hemodynamic loads is a particular challenge, given the risk of thrombosis and adverse remodeling that it brings. However, the blood is also a source of (immune) cells and proteins that dominantly contribute to functional tissue regeneration. This review explores the potential of the blood as a source for the complete or partial in situ regeneration of cardiovascular tissues, with a particular focus on the endothelium, being the natural blood-tissue barrier. We pinpoint the current scientific challenges to enable rational engineering and testing of blood-contacting implants to leverage the regenerative potential of the blood.
Collapse
Affiliation(s)
- Dina M Ibrahim
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Aleksandra Fomina
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Graduate School of Life Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
18
|
Gao Q, Jia F, Li X, Kong Y, Tian Z, Bi L, Li L. Biophysical cues to improve the immunomodulatory capacity of mesenchymal stem cells: The progress and mechanisms. Biomed Pharmacother 2023; 162:114655. [PMID: 37031489 DOI: 10.1016/j.biopha.2023.114655] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can maintain immune homeostasis and many preclinical trials with MSCs have been carried out around the world. In vitro culture of MSCs has been found to result in the decline of immunomodulatory capacity, migration and proliferation. To address these problems, simulating the extracellular environment for preconditioning of MSCs is a promising and inexpensive method. Biophysical cues in the external environment that MSCs are exposed to have been shown to affect MSC migration, residency, differentiation, secretion, etc. We review the main ways in which MSCs exert their immunomodulatory ability, and summarize recent advances in mechanical preconditioning of MSCs to enhance immunomodulatory capacity and related mechanical signal sensing and transduction mechanisms.
Collapse
Affiliation(s)
- Qingyuan Gao
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Fangru Jia
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Xiangpan Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Yanan Kong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Zhenya Tian
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China.
| |
Collapse
|
19
|
Impact of Non-Pharmacological Interventions on the Mechanisms of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23169097. [PMID: 36012362 PMCID: PMC9409393 DOI: 10.3390/ijms23169097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis remains the leading cause of mortality and morbidity worldwide characterized by the deposition of lipids and fibrous elements in the form of atheroma plaques in vascular areas which are hemodynamically overloaded. The global burden of atherosclerotic cardiovascular disease is steadily increasing and is considered the largest known non-infectious pandemic. The management of atherosclerotic cardiovascular disease is increasing the cost of health care worldwide, which is a concern for researchers and physicians and has caused them to strive to find effective long-term strategies to improve the efficiency of treatments by managing conventional risk factors. Primary prevention of atherosclerotic cardiovascular disease is the preferred method to reduce cardiovascular risk. Fasting, a Mediterranean diet, and caloric restriction can be considered useful clinical tools. The protective impact of physical exercise over the cardiovascular system has been studied in recent years with the intention of explaining the mechanisms involved; the increase in heat shock proteins, antioxidant enzymes and regulators of cardiac myocyte proliferation concentration seem to be the molecular and biochemical shifts that are involved. Developing new therapeutic strategies such as vagus nerve stimulation, either to prevent or slow the disease’s onset and progression, will surely have a profound effect on the lives of millions of people.
Collapse
|
20
|
Cabiati M, Giacomarra M, Fontanini M, Cecchettini A, Pelosi G, Vozzi F, Del Ry S. Bone morphogenetic protein-4 system expression in human coronary artery endothelial and smooth muscle cells under dynamic flow: effect of medicated bioresorbable vascular scaffolds at low and normal shear stress. Heart Vessels 2022; 37:2137-2149. [PMID: 35857064 DOI: 10.1007/s00380-022-02140-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/06/2022] [Indexed: 11/04/2022]
Abstract
Endothelial and smooth muscle cell dysfunction is an early event at the onset of atherosclerosis, a heterogeneous and multifactorial pathology of the vascular wall. Bone morphogenetic protein (BMP)-4, a mechanosensitive autocrine cytokine, and BMPR-1a, BMPR-1b, BMPR-2 specific receptors play a key role in atherosclerotic plaque formation and vascular calcification and BMP4 is regarded as a biomarker of endothelial cell activation. The study aimed to examine the BMP4 system expression by Real-Time PCR in Human Coronary Artery Endothelial (HCAECs) and Smooth Muscle Cells (HCASMCs) under different flow rates determining low or physiological shear stress in the presence/absence of medicated Bioresorbable Vascular Scaffold (BVS). The HCAEC and HCASMC were subjected to 1-10-20 dyne/cm2 shear stress in a laminar flow bioreactor system, with/without BVS+ Everolimus (600 nM). In HCAECs without BVS the BMP4 expression was similar at 1, 20 dyne/cm2 decreasing at 10 dyne/cm2, while adding BVS+ Everolimus, it decreased both at 1, 10 compared to 20 dyne/cm2. In HCASMCs without BVS + Everolimus, the BMP4 system mRNA expression was significantly reduced at 1, 10 dyne/cm2 compared to 20 dyne/cm2, while in the presence of BVS+ Everolimus, higher BMP4 mRNA levels were observed at 10 compared to 1, 20 dyne/cm2. In HCAECs and HCASMCs BMPRs were expressed in all experimental conditions except for BMPR-1a at 1 dyne/cm2 in HCAEC. Significant correlations were found between BMP4 and BMPRs. The more negligible on BMP4 expression due to low shear stress in HCAEC compared to HCASMC and its reduction in the presence of BVS+ Everolimus at low shear stress highlighted the protection of BMP4-mediated against endothelial dysfunction and neoatherogenesis.
Collapse
Affiliation(s)
- Manuela Cabiati
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology CNR, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy.
| | - Manuel Giacomarra
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology CNR, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Martina Fontanini
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology CNR, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Antonella Cecchettini
- Laboratory of Proteomics, Institute of Clinical Physiology, IFC-CNR, Pisa, Italy.,Department of Experimental and Clinical Medicine, University of Pisa, Pisa, Italy
| | - Gualtiero Pelosi
- Laboratory of Biomimetic Materials and Tissue Engineering, Institute of Clinical Physiology CNR, Pisa, Italy
| | - Federico Vozzi
- Laboratory of Biomimetic Materials and Tissue Engineering, Institute of Clinical Physiology CNR, Pisa, Italy
| | - Silvia Del Ry
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology CNR, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
21
|
Munger MA, Nelson SD, Teng C, Cheung AK, Sauer BC. Reduced risk of
NSAID‐Induced
adverse events with concomitant use of misoprostol (
MICRO
study). Pharmacotherapy 2022; 42:540-548. [DOI: 10.1002/phar.2708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/20/2022] [Accepted: 05/25/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Mark A. Munger
- Department of Pharmacotherapy University of Utah Salt Lake City Utah USA
- Division of Cardiology, Department of Internal Medicine University of Utah Salt Lake City Utah USA
| | - Scott D. Nelson
- Department of Pharmacotherapy University of Utah Salt Lake City Utah USA
- Department of Biomedical Informatics Vanderbilt University Nashville Tennessee USA
| | - Chia‐Chen Teng
- SLC Veterans Affairs Informatics, Decision‐Enhancement and Analytic Sciences, Health Services Research and Development Center and Team VERITAS, Division of Epidemiology University of Utah Salt Lake City Utah USA
| | - Alfred K. Cheung
- Division of Nephrology and Hypertension, Department of Internal Medicine The Veterans Affairs Salt Lake City Healthcare System Salt Lake City Utah USA
| | - Brian C. Sauer
- SLC Veterans Affairs Informatics, Decision‐Enhancement and Analytic Sciences, Health Services Research and Development Center and Team VERITAS, Division of Epidemiology University of Utah Salt Lake City Utah USA
| |
Collapse
|
22
|
Salvador J, Iruela-Arispe ML. Nuclear Mechanosensation and Mechanotransduction in Vascular Cells. Front Cell Dev Biol 2022; 10:905927. [PMID: 35784481 PMCID: PMC9247619 DOI: 10.3389/fcell.2022.905927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Vascular cells are constantly subjected to physical forces associated with the rhythmic activities of the heart, which combined with the individual geometry of vessels further imposes oscillatory, turbulent, or laminar shear stresses on vascular cells. These hemodynamic forces play an important role in regulating the transcriptional program and phenotype of endothelial and smooth muscle cells in different regions of the vascular tree. Within the aorta, the lesser curvature of the arch is characterized by disturbed, oscillatory flow. There, endothelial cells become activated, adopting pro-inflammatory and athero-prone phenotypes. This contrasts the descending aorta where flow is laminar and endothelial cells maintain a quiescent and atheroprotective phenotype. While still unclear, the specific mechanisms involved in mechanosensing flow patterns and their molecular mechanotransduction directly impact the nucleus with consequences to transcriptional and epigenetic states. The linker of nucleoskeleton and cytoskeleton (LINC) protein complex transmits both internal and external forces, including shear stress, through the cytoskeleton to the nucleus. These forces can ultimately lead to changes in nuclear integrity, chromatin organization, and gene expression that significantly impact emergence of pathology such as the high incidence of atherosclerosis in progeria. Therefore, there is strong motivation to understand how endothelial nuclei can sense and respond to physical signals and how abnormal responses to mechanical cues can lead to disease. Here, we review the evidence for a critical role of the nucleus as a mechanosensor and the importance of maintaining nuclear integrity in response to continuous biophysical forces, specifically shear stress, for proper vascular function and stability.
Collapse
Affiliation(s)
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
23
|
Meng F, Cheng H, Qian J, Dai X, Huang Y, Fan Y. In vitro fluidic systems: Applying shear stress on endothelial cells. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
24
|
Tang SY, FitzGerald GA. Endothelial Tenascin-X Is Going With the Flow. Circ Res 2022; 130:1660-1661. [PMID: 35617358 DOI: 10.1161/circresaha.122.321196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Soon Yew Tang
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Philadelphia, PA
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Philadelphia, PA
| |
Collapse
|
25
|
Wilde C, Mitgau J, Suchý T, Schoeneberg T, Liebscher I. Translating the Force - mechano-sensing GPCRs. Am J Physiol Cell Physiol 2022; 322:C1047-C1060. [PMID: 35417266 DOI: 10.1152/ajpcell.00465.2021] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Incorporating mechanical cues into cellular responses allows us to experience our direct environment. Specialized cells can perceive and discriminate between different physical properties such as level of vibration, temperature, or pressure. Mechanical forces are abundant signals that also shape general cellular responses such as cytoskeletal rearrangement, differentiation, or migration and contribute to tissue development and function. The molecular structures that perceive and transduce mechanical forces are specialized cytoskeletal proteins, cell junction molecules, and membrane proteins such as ion channels and metabotropic receptors. G protein-coupled receptors (GPCRs) have attracted attention as metabotropic force receptors as they are among the most important drug targets. This review summarizes the function of mechano-sensitive GPCRs, specifically, the angiotensin II type 1 receptor and adrenergic, apelin, histamine, parathyroid hormone 1, and orphan receptors, focusing particularly on the advanced knowledge gained from adhesion-type GPCRs. We distinguish between shear stress and cell swelling/stretch as the two major types of mechano-activation of these receptors and contemplate the potential contribution of the force-from-lipid and force-from-tether models that have previously been suggested for ion channels.
Collapse
Affiliation(s)
- Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Jakob Mitgau
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Tomás Suchý
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Torsten Schoeneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| |
Collapse
|
26
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
27
|
Vascular Pathobiology: Atherosclerosis and Large Vessel Disease. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
28
|
Kulkarni R, Andraska E, McEnaney R. Structural Remodeling of the Extracellular Matrix in Arteriogenesis: A Review. Front Cardiovasc Med 2021; 8:761007. [PMID: 34805316 PMCID: PMC8602576 DOI: 10.3389/fcvm.2021.761007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/10/2023] Open
Abstract
Lower extremity arterial occlusive disease (AOD) results in significant morbidity and mortality for the population, with up to 10% of patients ultimately requiring amputation. An alternative method for non-surgical revascularization which is yet to be fully understood is the optimization of the body's own natural collateral arterial network in a process known as arteriogenesis. Under conditions of conductance vessel stenosis or occlusion resulting in increased flow, shear forces, and pressure gradients within collaterals, positive remodeling occurs to increase the diameter and capacity of these vessels. The creation of a distal arteriovenous fistula (AVF) will drive increased arteriogenesis as compared to collateral formation with the occlusion of a conductance vessel alone by further increasing flow through these arterioles, demonstrating the capacity for arteriogenesis to form larger, more efficient collaterals beyond what is spontaneously achieved after arterial occlusion. Arteries rely on an extracellular matrix (ECM) composed of elastic fibers and collagens that provide stability under hemodynamic stress, and ECM remodeling is necessary to allow for increased diameter and flow conductance in mature arterial structures. When positive remodeling occurs, digestion of lamella and the internal elastic lamina (IEL) by matrix metalloproteinases (MMPs) and other elastases results in the rearrangement and thinning of elastic structures and may be replaced with disordered elastin synthesis without recovery of elastic function. This results in transmission of wall strain to collagen and potential for aneurysmal degeneration along collateral networks, as is seen in the pancreaticoduodenal artery (PDA) after celiac occlusion and inferior mesenteric artery (IMA) with concurrent celiac and superior mesenteric artery (SMA) occlusions. Further understanding into the development of collaterals is required to both better understand aneurysmal degeneration and optimize collateral formation in AOD.
Collapse
Affiliation(s)
- Rohan Kulkarni
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Elizabeth Andraska
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ryan McEnaney
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Veterans Affairs Hospitals Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
29
|
Ishii T, Warabi E, Mann GE. Mechanisms underlying unidirectional laminar shear stress-mediated Nrf2 activation in endothelial cells: Amplification of low shear stress signaling by primary cilia. Redox Biol 2021; 46:102103. [PMID: 34425388 PMCID: PMC8379703 DOI: 10.1016/j.redox.2021.102103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells are sensitive to mechanical stress and respond differently to oscillatory flow versus unidirectional flow. This review highlights the mechanisms by which a wide range of unidirectional laminar shear stress induces activation of the redox sensitive antioxidant transcription factor nuclear factor-E2-related factor 2 (Nrf2) in cultured endothelial cells. We propose that fibroblast growth factor-2 (FGF-2), brain-derived neurotrophic factor (BDNF) and 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) are potential Nrf2 activators induced by laminar shear stress. Shear stress-dependent secretion of FGF-2 and its receptor-mediated signaling is tightly controlled, requiring neutrophil elastase released by shear stress, αvβ3 integrin and the cell surface glycocalyx. We speculate that primary cilia respond to low laminar shear stress (<10 dyn/cm2), resulting in secretion of insulin-like growth factor 1 (IGF-1), which facilitates αvβ3 integrin-dependent FGF-2 secretion. Shear stress induces generation of heparan-binding epidermal growth factor-like growth factor (HB-EGF), which contributes to FGF-2 secretion and gene expression. Furthermore, HB-EGF signaling modulates FGF-2-mediated NADPH oxidase 1 activation that favors casein kinase 2 (CK2)-mediated phosphorylation/activation of Nrf2 associated with caveolin 1 in caveolae. Higher shear stress (>15 dyn/cm2) induces vesicular exocytosis of BDNF from endothelial cells, and we propose that BDNF via the p75NTR receptor could induce CK2-mediated Nrf2 activation. Unidirectional laminar shear stress upregulates gene expression of FGF-2 and BDNF and generation of 15d-PGJ2, which cooperate in sustaining Nrf2 activation to protect endothelial cells against oxidative damage.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
30
|
Dual Versus Single Oxygenated Hypothermic Machine Perfusion of Porcine Livers: Impact on Hepatobiliary and Endothelial Cell Injury. Transplant Direct 2021; 7:e741. [PMID: 34386578 PMCID: PMC8354629 DOI: 10.1097/txd.0000000000001184] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/09/2021] [Indexed: 12/12/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. Hypothermic oxygenated machine perfusion (HOPE) reduces ischemia-reperfusion injury of donor livers and is increasingly used in clinical transplantation. However, it remains unclear whether perfusion via the portal vein alone (HOPE) or via both the portal vein and hepatic artery (dual HOPE or DHOPE) is superior. Methods. Twelve porcine livers donated after circulatory death were randomized for 2 h of HOPE (n = 6) or DHOPE (n = 6), followed by 4 h of warm reperfusion with whole blood, to mimic transplantation. Hepatobiliary and endothelial cell function and injury markers were determined in perfusate and bile samples. Biopsies of bile ducts, hepatic arteries, and liver parenchyma were collected to assess histological damage and the expression of endothelial protective genes (KLF-2, eNOS, ET-1, CD31, VWF, VEGF-A). Results. There were no differences in hepatobiliary function and injury after warm reperfusion between the groups, apart from a 2-fold lower concentration of alanine aminotransferase in the perfusate (P = 0.045) and a lower peak lactate dehydrogenase in bile (P = 0.04) of livers preserved by DHOPE. Endothelial cell function and injury, as assessed by perfusate nitric oxide and von Willebrand factor antigen levels, as well as endothelial protective gene expressions, were similar between the groups. The hepatic arteries of both groups showed no microscopic evidence of injury. Conclusions. This study did not reveal major differences in hepatobiliary or endothelial function and injury after preservation by single or dual HOPE of porcine livers donated after circulatory death.
Collapse
|
31
|
Chies AB, Spadella MA, de Oliveira PR, Domeniconi RF, de Mello Santos T, Moreira RP, Rosales CB, Casarini DE, Navar LG. Exercise-Induced Modulation of Angiotensin II Responses in Femoral Veins From 2-Kidney-1-Clip Hypertensive Rats. Front Physiol 2021; 12:620438. [PMID: 33897446 PMCID: PMC8058411 DOI: 10.3389/fphys.2021.620438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/15/2021] [Indexed: 11/17/2022] Open
Abstract
The present study investigated the angiotensin II (Ang II) responses in rat femoral veins taken from 2-kidney-1clip (2K1C) hypertensive rats at 4 weeks after clipping, as well as the effects of exercise on these responses. In this manner, femoral veins taken from 2K1C rats kept at rest or exposed to acute exercise or to exercise training were challenged with Ang II or endothelin-1 (ET-1) in organ bath. Simultaneously, the presence of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) were determined in these preparations by western blotting. In these experiments, femoral veins exhibited subdued Ang II responses. However, after nitric oxide (NO) synthesis blockade, the responses were higher in the femoral veins taken from animals kept at rest [0.137(0.049–0.245); n = 10] than those obtained in trained animals kept at rest [0.008(0.001–0.041); n = 10] or studied after a single bout of exercise [0.001(0.001–0.054); n = 11]. In preparations in which, in addition to NO synthesis, both the local production of prostanoids and the action of ET-1 on type A (ETA) or B (ETB) receptors were inhibited, the differences induced by exercise were no longer observed. In addition, neither ET-1 responses nor the presence of COX-1 and COX-2 in these preparations were modified by the employed exercise protocols. In conclusion, NO maintains Ang II responses reduced in femoral veins of 2K1C animals at rest. However, vasodilator prostanoids as well as other relaxing mechanisms, activated by ETB stimulation, are mobilized by exercise to cooperate with NO in order to maintain controlled Ang II responses in femoral veins.
Collapse
Affiliation(s)
| | | | | | | | | | - Roseli Peres Moreira
- Department of Medicine, Nephrology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla B Rosales
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States
| | - Dulce Elena Casarini
- Department of Medicine, Nephrology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luis Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
32
|
Yang X, Lu T, Qu Z, Zhang Y, Liu P, Ma Y. Plasma D-dimer level is associated with clinical outcomes in patients with atrial fibrillation related acute ischemic stroke after pneumonia. BMC Neurol 2021; 21:137. [PMID: 33773590 PMCID: PMC8004437 DOI: 10.1186/s12883-021-02168-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background Pneumonia is related to poor prognosis in acute ischemic stroke (AIS), and its risk might be higher in atrial fibrillation (AF) related AIS with elevated plasma D-dimer. The aim of our study was to investigate the prognostic value of D-dimer for predicting clinical outcome of AF-related AIS with pneumonia. Method AF-related AIS patients with pneumonia were prospectively enrolled. Receiver operating characteristic (ROC) curve was used to determine the optimal D-dimer point for 3-month mortality and death/severe disability. The associations between the D-dimer and 3-month mortality and death/severe disability were assessed by multivariable logistic regression analysis. Results A total of 415 patients were enrolled in this study. ROC curve analysis showed that the optimal cut point of D-dimer for 3-month death/severe disability and mortality were D-dimer≥2.35 mg/l and D-dimer≥3.35 mg/l, respectively. Multivariable logistic regression analysis showed that D-dimer≥2.35 mg/l [adjusted odds ratio (aOR) 5.99, 95% confidence interval (CI): 3.04–11.83, P<0.001], higher NIHISS score (aOR:1.53, 95% CI: 1.38–1.69, P<0.001) and larger infarct volume (aOR 1.01, 95% CI: 1.01–1.02, P<0.001) were associated with increased risk of 3-month death/severe disability), and anticoagulant was associated with decreased risk of death/severe disability (aOR:0.21, 95% CI: 0.09–0.47, P<0.001). Higher NIHISS score (aOR:1.64, 95% CI: 1.38–1.94, P<0.001), older age (aOR 1.08, 95% CI: 1.02–1.14, P = 0.007), D-dimer≥3.35 mg/l (OR 8.49, 95% CI: 4.13–17.84,P<0.001), larger infarct volume (aOR 1.02, 95% CI: 1.00–1.03, P = 0.014), and higher CRUB-65 score (aOR 6.43, 95% CI: 3.10–13.34, P<0.001) were associated with increased risk of 3-month mortality. Conclusions AF-related AIS patients with concurrent high D-dimer and pneumonia increased risk of 3-month mortality and death/severe disability, plasma D-dimer may have predictive value in outcome after AF-related AIS with pneumonia.
Collapse
Affiliation(s)
- Xu Yang
- Department of Neurology, Nanchong Central Hospital; The Second Clinical Medical School, North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Taoli Lu
- Department of Neurology, Chengdu second people's hospital, Chengdu, 610015, Sichuan, China
| | - Zhanli Qu
- Department of Neurology, Nanchong Central Hospital; The Second Clinical Medical School, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yi Zhang
- Department of Neurology, Nanchong Central Hospital; The Second Clinical Medical School, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Pingping Liu
- Department of Neurology, Nanchong Central Hospital; The Second Clinical Medical School, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Ying Ma
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| |
Collapse
|
33
|
Singh B, Kosuru R, Lakshmikanthan S, Sorci-Thomas M, Zhang D, Sparapani R, Vasquez-Vivar J, Chrzanowska M. Endothelial Rap1 (Ras-Association Proximate 1) Restricts Inflammatory Signaling to Protect From the Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:638-650. [PMID: 33267664 PMCID: PMC8105264 DOI: 10.1161/atvbaha.120.315401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Small GTPase Rap1 (Ras-association proximate 1) is a novel, positive regulator of NO release and endothelial function with a potentially key role in mechanosensing of atheroprotective, laminar flow. Our objective was to delineate the role of Rap1 in the progression of atherosclerosis and its specific functions in the presence and absence of laminar flow, to better define its role in endothelial mechanisms contributing to plaque formation and atherogenesis. Approach and Results: In a mouse atherosclerosis model, endothelial Rap1B deletion exacerbates atherosclerotic plaque formation. In the thoracic aorta, where laminar shear stress-induced NO is otherwise atheroprotective, plaque area is increased in Athero-Rap1BiΔEC (atherogenic endothelial cell-specific, tamoxifen-inducible Rap1A+Rap1B knockout) mice. Endothelial Rap1 deficiency also leads to increased plaque size, leukocyte accumulation, and increased CAM (cell adhesion molecule) expression in atheroprone areas, whereas vascular permeability is unchanged. In endothelial cells, in the absence of protective laminar flow, Rap1 deficiency leads to an increased proinflammatory TNF-α (tumor necrosis factor alpha) signaling and increased NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation and elevated inflammatory receptor expression. Interestingly, this increased signaling to NF-κB activation is corrected by AKTVIII-an inhibitor of Akt (protein kinase B) translocation to the membrane. Together, these data implicate Rap1 in restricting Akt-dependent signaling, preventing excessive cytokine receptor signaling and proinflammatory NF-κB activation. CONCLUSIONS Via 2 distinct mechanisms, endothelial Rap1 protects from the atherosclerosis progression in the presence and absence of laminar flow; Rap1-stimulated NO release predominates in laminar flow, and restriction of proinflammatory signaling predominates in the absence of laminar flow. Our studies provide novel insights into the mechanisms underlying endothelial homeostasis and reveal the importance of Rap1 signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Bandana Singh
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | - Ramoji Kosuru
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | | | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Endocrinology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rodney Sparapani
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeannette Vasquez-Vivar
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Magdalena Chrzanowska
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
34
|
Badimon L, Vilahur G, Rocca B, Patrono C. The key contribution of platelet and vascular arachidonic acid metabolism to the pathophysiology of atherothrombosis. Cardiovasc Res 2021; 117:2001-2015. [PMID: 33484117 DOI: 10.1093/cvr/cvab003] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Arachidonic acid is one of the most abundant and ubiquitous ω-6 polyunsaturated fatty acid, present in esterified form in the membrane phospholipids of all mammalian cells and released from phospholipids by several phospholipases in response to various activating or inhibitory stimuli. Arachidonic acid is the precursor of a large number of enzymatically and non-enzymatically derived, biologically active autacoids, including prostaglandins (PGs), thromboxane (TX) A2, leukotrienes, and epoxyeicosatetraenoic acids (collectively called eicosanoids), endocannabinoids and isoprostanes, respectively. Eicosanoids are local modulators of the physiological functions and pathophysiological roles of blood vessels and platelets. For example, the importance of cyclooxygenase (COX)-1-derived TXA2 from activated platelets in contributing to primary haemostasis and atherothrombosis is demonstrated in animal and human models by the bleeding complications and cardioprotective effects associated with low-dose aspirin, a selective inhibitor of platelet COX-1. The relevance of vascular COX-2-derived prostacyclin (PGI2) in endothelial thromboresistance and atheroprotection is clearly shown by animal and human models and by the adverse cardiovascular effects exerted by COX-2 inhibitors in humans. A vast array of arachidonic acid-transforming enzymes, downstream synthases and isomerases, transmembrane receptors, and specificity in their tissue expression make arachidonic acid metabolism a fine-tuning system of vascular health and disease. Its pharmacological regulation is central in human cardiovascular diseases, as demonstrated by biochemical measurements and intervention trials.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Bianca Rocca
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy.,Gemelli' Foundation, IRCCS, Rome, Italy
| | - Carlo Patrono
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy.,Gemelli' Foundation, IRCCS, Rome, Italy
| |
Collapse
|
35
|
Hirata T, Yamamoto K, Ikeda K, Arita M. Functional lipidomics of vascular endothelial cells in response to laminar shear stress. FASEB J 2021; 35:e21301. [PMID: 33421194 DOI: 10.1096/fj.202002144r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 11/11/2022]
Abstract
Laminar shear stress generated by blood flow stimulates endothelial cells and activates signal transduction, which plays an important role in vascular homeostasis. Several lines of evidence indicate that membrane and intracellular lipids are involved in the signal transduction of biomechanical stresses. In this study, we performed global profiling of cellular lipids from human pulmonary artery endothelial cells (HPAEC) exposed to laminar shear stress. A total of 761 species of lipids were successfully annotated, with 198 of these species significantly changed in response to shear stress for 24 hours. Ether-linked lipids containing an alkyl moiety with a medium chain length (C11-C14) were uniquely upregulated, and the administration of their biosynthetic precursor 1-O-dodecyl-rac-glycerol attenuated phorbol 12-myristate 13-acetate (PMA) induced vascular cell adhesion molecule-1 (VCAM-1) expression. Given the pro-inflammatory and atherogenic roles of VCAM-1, our findings suggest that the induction of a specific group of lipids (ie, ether-linked lipids with medium length alkyl side chain) may confer atheroprotective and anti-inflammatory roles to vascular endothelial cells under flow conditions.
Collapse
Affiliation(s)
- Tsuyoshi Hirata
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory of Biomolecule Analysis, Kazusa DNA Research Institute, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| |
Collapse
|
36
|
Alali MH, Vianna LC, Lucas RAI, Junejo RT, Fisher JP. Impact of whole body passive heat stress and arterial shear rate modification on radial artery function in young men. J Appl Physiol (1985) 2020; 129:1373-1382. [PMID: 33031019 DOI: 10.1152/japplphysiol.00296.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We sought to determine how whole body heating acutely influences radial artery function, characterized using flow-mediated dilation (FMD) and low-flow-mediated constriction (L-FMC), and the mechanistic role of shear rate modification on radial artery functional characteristics during heating. Eleven young healthy men underwent whole body heating (water-perfused suit) sufficient to raise the core temperature by +1°C. Trials were repeated with (heat + WC) and without (heat) the application of a wrist cuff located distal to the radial artery examined, known to prevent increases in mean and anterograde shear rates but increase retrograde shear rate. Radial artery characteristics were assessed throughout each trial, with FMD and L-FMC assessed before and upon reaching the target core temperature. Heat markedly increased radial artery mean and anterograde shear rates, along with radial artery diameter and blood flow (P < 0.05). Heat + WC abolished the heat-induced increase in mean and anterograde shear rates (P > 0.05) but markedly increased retrograde shear rate (P < 0.05). Concomitantly, increases in radial artery diameter and blood flow were decreased (heat + WC vs. heat, P < 0.05). Heat attenuated FMD (8.6 ± 1.2% vs. 2.2 ± 1.4%, P < 0.05), whereas no change in FMD was observed in heat + WC (7.8 ± 1.2% vs. 10.8 ± 1.2%, P > 0.05). In contrast, L-FMC was not different in either trial (P > 0.05). In summary, acute whole body heating markedly elevates radial artery shear rate, diameter, and blood flow and diminishes FMD. However, marked radial artery vasodilation and diminished FMD are absent when these shear rate changes are prevented. Shear rate modifications underpin the radial artery response to acute whole body heat stress, but further endothelium-dependent vasodilation (FMD) is attenuated likely as the vasodilatory range limit is approached.NEW & NOTEWORTHY We observed that acute whole body heating elevates radial artery shear rate, diameter, and blood flow. This results in a diminished flow-meditated dilatation (FMD) but does not change low-flow-mediated constriction (L-FMC). Preventing shear rate changes during whole body heating reduces radial artery vasodilation and reverses FMD reductions but has no effect on L-FMC. These findings indicate that shear rate changes underpin conduit artery responses to acute whole body heat stress, but further endothelium-dependent flow-mediated vasodilation is attenuated as the vasodilatory range limit is approached.
Collapse
Affiliation(s)
- Mohammad H Alali
- School of Sport, Exercise & Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lauro C Vianna
- NeuroV̇ASQ̇ - Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasília, Distrito Federal, Brazil
| | - Rebekah A I Lucas
- School of Sport, Exercise & Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rehan T Junejo
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom.,Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, United Kingdom
| | - James P Fisher
- Faculty of Medical & Health Sciences, Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
37
|
Bonezzi C, Fornasari D, Cricelli C, Magni A, Ventriglia G. Pharmacological Management of Adults with Chronic Non-Cancer Pain in General Practice. Pain Ther 2020; 9:17-28. [PMID: 33315207 PMCID: PMC7736454 DOI: 10.1007/s40122-020-00218-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022] Open
Abstract
Chronic pain is a public health priority that affects about 20% of the general population, causing disability and impacting productivity and quality of life. It is often managed in the primary care setting. Chronic pain management is most effective when the pain mechanism has been identified and addressed by appropriate therapy. This document provides an overview of pharmacological therapy for chronic non-cancer pain in the primary care setting, with the aim of improving treatment decisions based on the underlying pain mechanisms and pain neuroscience.
Collapse
Affiliation(s)
- Cesare Bonezzi
- ICS Maugeri IRCCS, Via Salvatore Maugeri 10, Pavia, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi Di Milano, Via Vanvitelli 32, Milan, Italy.
| | - Claudio Cricelli
- SIMG (Italian College of General Practitioners and Primary Care), Via Del Sansovino 179, Florence, Italy
| | - Alberto Magni
- SIMG (Italian College of General Practitioners and Primary Care), Via Del Sansovino 179, Florence, Italy
| | - Giuseppe Ventriglia
- SIMG (Italian College of General Practitioners and Primary Care), Via Del Sansovino 179, Florence, Italy
| |
Collapse
|
38
|
Zaric BL, Radovanovic JN, Gluvic Z, Stewart AJ, Essack M, Motwalli O, Gojobori T, Isenovic ER. Atherosclerosis Linked to Aberrant Amino Acid Metabolism and Immunosuppressive Amino Acid Catabolizing Enzymes. Front Immunol 2020; 11:551758. [PMID: 33117340 PMCID: PMC7549398 DOI: 10.3389/fimmu.2020.551758] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the leading global health concern and responsible for more deaths worldwide than any other type of disorder. Atherosclerosis is a chronic inflammatory disease in the arterial wall, which underpins several types of cardiovascular disease. It has emerged that a strong relationship exists between alterations in amino acid (AA) metabolism and the development of atherosclerosis. Recent studies have reported positive correlations between levels of branched-chain amino acids (BCAAs) such as leucine, valine, and isoleucine in plasma and the occurrence of metabolic disturbances. Elevated serum levels of BCAAs indicate a high cardiometabolic risk. Thus, BCAAs may also impact atherosclerosis prevention and offer a novel therapeutic strategy for specific individuals at risk of coronary events. The metabolism of AAs, such as L-arginine, homoarginine, and L-tryptophan, is recognized as a critical regulator of vascular homeostasis. Dietary intake of homoarginine, taurine, and glycine can improve atherosclerosis by endothelium remodeling. Available data also suggest that the regulation of AA metabolism by indoleamine 2,3-dioxygenase (IDO) and arginases 1 and 2 are mediated through various immunological signals and that immunosuppressive AA metabolizing enzymes are promising therapeutic targets against atherosclerosis. Further clinical studies and basic studies that make use of animal models are required. Here we review recent data examining links between AA metabolism and the development of atherosclerosis.
Collapse
Affiliation(s)
- Bozidarka L. Zaric
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena N. Radovanovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran Gluvic
- Department of Endocrinology and Diabetes, Faculty of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, University of Belgrade, Belgrade, Serbia
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Magbubah Essack
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Olaa Motwalli
- College of Computing and Informatics, Saudi Electronic University (SEU), Medina, Saudi Arabia
| | - Takashi Gojobori
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
39
|
Junejo RT, Lip GYH, Fisher JP. Cerebrovascular Dysfunction in Atrial Fibrillation. Front Physiol 2020; 11:1066. [PMID: 33013456 PMCID: PMC7509200 DOI: 10.3389/fphys.2020.01066] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/04/2020] [Indexed: 01/13/2023] Open
Abstract
It is now well established that besides being the most common sustained arrhythmia, atrial fibrillation (AF) is a major healthcare burden. Risk of debilitating stroke is increased in AF patients, but even in the absence of stroke, this population is at heightened risk of cognitive decline, depression, and dementia. The reasons for this are complex, multifactorial, and incompletely understood. One potential contributing mechanism is cerebrovascular dysfunction. Cerebral blood flow is regulated by chemical, metabolic, autoregulatory, neurogenic, and systemic factors. The dysfunction in one or more of these mechanisms may contribute to the elevated risk of cognitive decline and cerebrovascular events in AF. This short review presents the evidence for diminished cerebral blood flow, cerebrovascular carbon dioxide reactivity (i.e., cerebrovascular vasodilatory reserve), cerebral autoregulation, and neurovascular coupling in AF patients when compared to control participants in sinus rhythm. Further work is needed to understand the physiological mechanisms underpinning these observations and their clinical significance in atrial fibrillation patients.
Collapse
Affiliation(s)
- Rehan T. Junejo
- Liverpool Centre for Cardiovascular Science, Liverpool Heart and Chest Hospital, University of Liverpool, Liverpool, United Kingdom
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, Liverpool Heart and Chest Hospital, University of Liverpool, Liverpool, United Kingdom
| | - James P. Fisher
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
40
|
Junejo RT, Braz ID, Lucas SJ, van Lieshout JJ, Phillips AA, Lip GY, Fisher JP. Neurovascular coupling and cerebral autoregulation in atrial fibrillation. J Cereb Blood Flow Metab 2020; 40:1647-1657. [PMID: 31426699 PMCID: PMC7370373 DOI: 10.1177/0271678x19870770] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The risk of cognitive decline and stroke is increased by atrial fibrillation (AF). We sought to determine whether neurovascular coupling and cerebral autoregulation are blunted in people with AF in comparison with age-matched, patients with hypertension and healthy controls. Neurovascular coupling was assessed using five cycles of visual stimulation for 30 s followed by 30 s with both eyes-closed. Cerebral autoregulation was examined using a sit-stand test, and a repeated squat-to-stand (0.1 Hz) manoeuvre with transfer function analysis of mean arterial pressure (MAP; input) and middle cerebral artery mean blood flow velocity (MCA Vm; output) relationships at 0.1 Hz. Visual stimulation increased posterior cerebral artery conductance, but the magnitude of the response was blunted in patients with AF (18 [8] %; mean [SD]) and hypertension (17 [8] %), in comparison with healthy controls (26 [9] %) (P < 0.05). In contrast, transmission of MAP to MCA Vm was greater in AF patients compared to hypertension and healthy controls, indicating diminished cerebral autoregulation. We have shown for the first time that AF patients have impaired neurovascular coupling responses to visual stimulation and diminished cerebral autoregulation. Such deficits in cerebrovascular regulation may contribute to the increased risk of cerebral dysfunction in people with AF.
Collapse
Affiliation(s)
- Rehan T Junejo
- School of Sport, Exercise & Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Birmingham, UK.,Liverpool Centre for Cardiovascular Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Igor D Braz
- Medical School, University Center of Volta Redonda, Volta Redonda, Brazil
| | - Samuel Je Lucas
- School of Sport, Exercise & Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Birmingham, UK.,Centre for Human Brain Health, College of Life & Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Johannes J van Lieshout
- Department of Internal Medicine, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Clinical Cardiovascular Physiology, AMC Center for Heart Failure Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,MRC/Arthritis Research UK Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - Aaron A Phillips
- Departments of Physiology, Pharmacology & Clinical Neurosciences, Libin Cardiovascular Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Gregory Yh Lip
- Liverpool Centre for Cardiovascular Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - James P Fisher
- School of Sport, Exercise & Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Birmingham, UK.,Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
41
|
Targeted anti-inflammatory therapy is a new insight for reducing cardiovascular events: A review from physiology to the clinic. Life Sci 2020; 253:117720. [PMID: 32360620 DOI: 10.1016/j.lfs.2020.117720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022]
Abstract
Despite considerable progressions, cardiovascular disease (CVD) is still one of the major causes of mortality around the world, indicates an important and unmet clinical need. Recently, extensive studies have been performed on the role of inflammatory factors as either a major or surrogate factor in the pathophysiology of CVD. Epidemiological observations suggest the theory of the role of inflammatory mediators in the development of cardiovascular events. This may support the idea that targeted anti-inflammatory therapies, on the background of traditional validated medical therapies, can play a significant role in prevention and even reduction of cardiovascular disorders. Many randomized controlled trials have shown that drugs commonly useful for primary and secondary prevention of CVD have an anti-inflammatory mechanism. Further, many anti-inflammatory drugs are being examined because of their potential to reduce the risk of cardiovascular problems. In this study, we review the process of inflammation in the development of cardiovascular events, both in vivo and clinical evidence in immunotherapy for CVD.
Collapse
|
42
|
Roche-Molina M, Hardwick B, Sanchez-Ramos C, Sanz-Rosa D, Gewert D, Cruz FM, Gonzalez-Guerra A, Andres V, Palma JA, Ibanez B, Mckenzie G, Bernal JA. The pharmaceutical solvent N-methyl-2-pyrollidone (NMP) attenuates inflammation through Krüppel-like factor 2 activation to reduce atherogenesis. Sci Rep 2020; 10:11636. [PMID: 32669659 PMCID: PMC7363918 DOI: 10.1038/s41598-020-68350-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/19/2020] [Indexed: 12/25/2022] Open
Abstract
N-methyl-2-pyrrolidone (NMP) is a versatile water-miscible polar aprotic solvent. It is used as a drug solubilizer and penetration enhancer in human and animal, yet its bioactivity properties remain elusive. Here, we report that NMP is a bioactive anti-inflammatory compound well tolerated in vivo, that shows efficacy in reducing disease in a mouse model of atherosclerosis. Mechanistically, NMP increases the expression of the transcription factor Kruppel-like factor 2 (KLF2). Monocytes and endothelial cells treated with NMP express increased levels of KLF2, produce less pro-inflammatory cytokines and adhesion molecules. We found that NMP attenuates monocyte adhesion to endothelial cells inflamed with tumor necrosis factor alpha (TNF-α) by reducing expression of adhesion molecules. We further show using KLF2 shRNA that the inhibitory effect of NMP on endothelial inflammation and subsequent monocyte adhesion is KLF2 dependent. Enhancing KLF2 expression and activity improves endothelial function, controls multiple genes critical for inflammation, and prevents atherosclerosis. Our findings demonstrate a consistent effect of NMP upon KLF2 activation and inflammation, biological processes central to atherogenesis. Our data suggest that inclusion of bioactive solvent NMP in pharmaceutical compositions to treat inflammatory disorders might be beneficial and safe, in particular to treat diseases of the vascular system, such as atherosclerosis.
Collapse
Affiliation(s)
- Marta Roche-Molina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain
| | - Bryn Hardwick
- MRC Cancer Unit At the University of Cambridge, Hutchison/MRC Research Centre, Box 197, Biomedical Campus, Hills Road, Cambridge, CB2 0XZ, UK
| | - Cristina Sanchez-Ramos
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain
| | - David Sanz-Rosa
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain.,CIBERCV, Madrid, Spain.,Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - Dirk Gewert
- DG Bioconsult Ltd, 50 Gilbert Road, Cambridge, CB4 3PE, UK
| | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain
| | - Andres Gonzalez-Guerra
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain
| | - Vicente Andres
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Joaquin A Palma
- Department of Development, Grupo STIG, Velázquez 11, 28001, Madrid, CP, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain.,CIBERCV, Madrid, Spain.,IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Grahame Mckenzie
- MRC Cancer Unit At the University of Cambridge, Hutchison/MRC Research Centre, Box 197, Biomedical Campus, Hills Road, Cambridge, CB2 0XZ, UK.
| | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro 3, CP28029, Madrid, Spain. .,CIBERCV, Madrid, Spain.
| |
Collapse
|
43
|
Gao P, Chen C, Howell ED, Li Y, Tober J, Uzun Y, He B, Gao L, Zhu Q, Siekmann AF, Speck NA, Tan K. Transcriptional regulatory network controlling the ontogeny of hematopoietic stem cells. Genes Dev 2020; 34:950-964. [PMID: 32499402 PMCID: PMC7328518 DOI: 10.1101/gad.338202.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Abstract
In this study from Gao et al., the authors performed RNA-seq and histone mark ChIP-seq to define the transcriptomes and epigenomes of cells representing key developmental stages of HSC ontogeny in mice. Using a novel computational algorithm, target inference via physical connection (TIPC), they constructed developmental stage-specific transcriptional regulatory networks by linking enhancers and predicted bound transcription factors to their target promoters, thus providing a useful resource for uncovering regulators of HSC formation. Hematopoietic stem cell (HSC) ontogeny is accompanied by dynamic changes in gene regulatory networks. We performed RNA-seq and histone mark ChIP-seq to define the transcriptomes and epigenomes of cells representing key developmental stages of HSC ontogeny in mice. The five populations analyzed were embryonic day 10.5 (E10.5) endothelium and hemogenic endothelium from the major arteries, an enriched population of prehematopoietic stem cells (pre-HSCs), fetal liver HSCs, and adult bone marrow HSCs. Using epigenetic signatures, we identified enhancers for each developmental stage. Only 12% of enhancers are primed, and 78% are active, suggesting the vast majority of enhancers are established de novo without prior priming in earlier stages. We constructed developmental stage-specific transcriptional regulatory networks by linking enhancers and predicted bound transcription factors to their target promoters using a novel computational algorithm, target inference via physical connection (TIPC). TIPC predicted known transcriptional regulators for the endothelial-to-hematopoietic transition, validating our overall approach, and identified putative novel transcription factors, including the broadly expressed transcription factors SP3 and MAZ. Finally, we validated a role for SP3 and MAZ in the formation of hemogenic endothelium. Our data and computational analyses provide a useful resource for uncovering regulators of HSC formation.
Collapse
Affiliation(s)
- Peng Gao
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Changya Chen
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Elizabeth D Howell
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Graduate Group in Cell and Molecular Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Yan Li
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Yasin Uzun
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Bing He
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Long Gao
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Qin Zhu
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Arndt F Siekmann
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Nancy A Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kai Tan
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
44
|
The quest for effective pharmacological suppression of neointimal hyperplasia. Curr Probl Surg 2020; 57:100807. [PMID: 32771085 DOI: 10.1016/j.cpsurg.2020.100807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
|
45
|
Maida CD, Vasto S, Di Raimondo D, Casuccio A, Vassallo V, Daidone M, Del Cuore A, Pacinella G, Cirrincione A, Simonetta I, Della Corte V, Rizzica S, Geraci G, Tuttolomondo A, Pinto A. Inflammatory activation and endothelial dysfunction markers in patients with permanent atrial fibrillation: a cross-sectional study. Aging (Albany NY) 2020; 12:8423-8433. [PMID: 32364529 PMCID: PMC7244079 DOI: 10.18632/aging.103149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
In recent years a growing body of evidence supported the role of inflammation in the initiation, maintenance and outcome of atrial fibrillation (AF). Nevertheless, despite a large amount of information, whether AF or the underlying structural heart disease (SHD) is the cause of the inflammatory process is still under debate. We, therefore, sought to determine if the inflammatory process reflect an underlying disease or the arrhythmia ‘per se’. We evaluated plasma levels of soluble Interleukin 2 Receptor Alpha (sIL-2Rα), TNF-α and IL-18 in 100 consecutive patients with permanent AF, (43 with a SHD and 57 without a SHD) compared to 121 age and sex-matched controls which had normal sinus rhythm. We also evaluated the endothelial function in both groups of patients using reactive hyperemia index (RHI) values measured by Endo-PAT2000. Compared to controls, AF patients showed higher circulating levels of inflammatory markers and a lower mean value of RHI. At multiple logistic regression analysis, the inflammatory markers and RHI were significantly associated with AF presence, whereas ROC curve analysis had good sensitivity and specificity in inflammatory variables and RHI for AF presence. No significant association was observed in the group of permanent AF patients, between inflammatory markers and the presence of an underlying SHD. These findings could help to clarify the role of inflammation in subjects with AF and suggest that the markers of systemic inflammation are not associated with the underlying cardiovascular disease, rather with the atrial fibrillation ‘per se’.
Collapse
Affiliation(s)
- Carlo Domenico Maida
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy.,Molecular and Clinical Medicine PhD Programme, University of Palermo, Palermo, Italy
| | - Sonya Vasto
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Palermo, Italy
| | - Domenico Di Raimondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Alessandra Casuccio
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro"Section of Public Health Epidemiology and Preventive Medicine, University of Palermo, Palermo, Italy
| | - Valerio Vassallo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Mario Daidone
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Alessandro Del Cuore
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Gaetano Pacinella
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Anna Cirrincione
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Irene Simonetta
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | | | - Salvatore Rizzica
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Giulio Geraci
- Unit of Nephrology and Hypertension, European Society of Hypertension Excellence Center, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Antonio Pinto
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|
46
|
Abstract
Prostanoids (prostaglandins, prostacyclin and thromboxane) belong to the oxylipin family of biologically active lipids generated from arachidonic acid (AA). Protanoids control numerous physiological and pathological processes. Cyclooxygenase (COX) is a rate-limiting enzyme involved in the conversion of AA into prostanoids. There are two COX isozymes: the constitutive COX-1 and the inducible COX-2. COX-1 and COX-2 have similar structures, catalytic activities, and subcellular localizations but differ in patterns of expression and biological functions. Non-selective COX-1/2 or traditional, non-steroidal anti-inflammatory drugs (tNSAIDs) target both COX isoforms and are widely used to relieve pain, fever and inflammation. However, the use of NSAIDs is associated with various side effects, particularly in the gastrointestinal tract. NSAIDs selective for COX-2 inhibition (coxibs) were purposefully designed to spare gastrointestinal toxicity, but predisposed patients to increased cardiovascular risks. These health complications from NSAIDs prompted interest in the downstream effectors of the COX enzymes as novel drug targets. This chapter describes various safety issues with tNSAIDs and coxibs, and discusses the current development of novel classes of drugs targeting the prostanoid pathway, including nitrogen oxide- and hydrogen sulfide-releasing NSAIDs, inhibitors of prostanoid synthases, dual inhibitors, and prostanoid receptor agonists and antagonists.
Collapse
|
47
|
Driver B, Marks DC, van der Wal DE. Not all (N)SAID and done: Effects of nonsteroidal anti-inflammatory drugs and paracetamol intake on platelets. Res Pract Thromb Haemost 2020; 4:36-45. [PMID: 31989083 PMCID: PMC6971311 DOI: 10.1002/rth2.12283] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/07/2019] [Accepted: 10/19/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are key mediators of hemostasis and thrombosis and can be inhibited by nonsteroidal anti-inflammatory drugs (NSAIDs). As a result, platelet donors are temporarily deferred from donating if they have recently taken NSAIDs such as aspirin or ibuprofen. Despite these measures, a proportion of platelet donations show exposure to these drugs; however, little is known about the effect of NSAIDs and their metabolites on platelet quality in vivo and during storage. In this review, the effect of NSAIDs on platelet function is summarized, with a focus on the widely consumed over-the-counter (OTC) medications aspirin, ibuprofen, and the non-NSAID paracetamol. Aspirin and ibuprofen have well-defined antiplatelet effects. In comparison, studies regarding the effect of paracetamol on platelets report variable findings. The timing and order of NSAID intake is important, as concurrent NSAID use can inhibit or potentiate platelet activation depending on the drug taken. NSAID deferral periods and maximum platelet shelf-life is set by each country and are revised regularly. Reduced donor deferral periods and longer platelet storage times may affect the quality of platelet products, and it is therefore important to identify the possible impact of NSAID intake on platelet quality before and after storage.
Collapse
Affiliation(s)
- Ben Driver
- Research and DevelopmentAustralian Red Cross Blood ServiceSydneyNSWAustralia
| | - Denese C. Marks
- Research and DevelopmentAustralian Red Cross Blood ServiceSydneyNSWAustralia
- Sydney Medical SchoolThe University of SydneySydneyNSWAustralia
| | | |
Collapse
|
48
|
Darçot E, Colotti R, Pellegrin M, Wilson A, Siegert S, Bouzourene K, Yerly J, Mazzolai L, Stuber M, van Heeswijk RB. Towards Quantification of Inflammation in Atherosclerotic Plaque in the Clinic - Characterization and Optimization of Fluorine-19 MRI in Mice at 3 T. Sci Rep 2019; 9:17488. [PMID: 31767900 PMCID: PMC6877590 DOI: 10.1038/s41598-019-53905-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
Fluorine-19 (19F) magnetic resonance imaging (MRI) of injected perfluorocarbons (PFCs) can be used for the quantification and monitoring of inflammation in diseases such as atherosclerosis. To advance the translation of this technique to the clinical setting, we aimed to 1) demonstrate the feasibility of quantitative 19F MRI in small inflammation foci on a clinical scanner, and 2) to characterize the PFC-incorporating leukocyte populations and plaques. To this end, thirteen atherosclerotic apolipoprotein-E-knockout mice received 2 × 200 µL PFC, and were scanned on a 3 T clinical MR system. 19F MR signal was detected in the aortic arch and its branches in all mice, with a signal-to-noise ratio of 11.1 (interquartile range IQR = 9.5–13.1) and a PFC concentration of 1.15 mM (IQR = 0.79–1.28). Imaging flow cytometry was used on another ten animals and indicated that PFC-labeled leukocytes in the aortic arch and it branches were mainly dendritic cells, macrophages and neutrophils (ratio 9:1:1). Finally, immunohistochemistry analysis confirmed the presence of those cells in the plaques. We thus successfully used 19F MRI for the noninvasive quantification of PFC in atherosclerotic plaque in mice on a clinical scanner, demonstrating the feasibility of detecting very small inflammation foci at 3 T, and advancing the translation of 19F MRI to the human setting.
Collapse
Affiliation(s)
- Emeline Darçot
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Roberto Colotti
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Maxime Pellegrin
- Division of Angiology, Heart and Vessel Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Anne Wilson
- Flow Cytometry Facility, Department of Formation and Research, University of Lausanne (UNIL), Epalinges, Switzerland
| | - Stefanie Siegert
- Flow Cytometry Facility, Department of Formation and Research, University of Lausanne (UNIL), Epalinges, Switzerland
| | - Karima Bouzourene
- Division of Angiology, Heart and Vessel Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jérôme Yerly
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Center for Biomedical Imaging (CIBM), Lausanne and Geneva, Switzerland
| | - Lucia Mazzolai
- Division of Angiology, Heart and Vessel Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Matthias Stuber
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Center for Biomedical Imaging (CIBM), Lausanne and Geneva, Switzerland
| | - Ruud B van Heeswijk
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
49
|
Raees A, Bakhamis A, Mohamed-Ali V, Bashah M, Al-Jaber M, Abraham D, Clapp LH, Orie NN. Altered cyclooxygenase-1 and enhanced thromboxane receptor activities underlie attenuated endothelial dilatory capacity of omental arteries in obesity. Life Sci 2019; 239:117039. [PMID: 31704447 DOI: 10.1016/j.lfs.2019.117039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 01/05/2023]
Abstract
AIMS Obesity is a risk factor for endothelial dysfunction, the severity of which is likely to vary depending on extent and impact of adiposity on the vasculature. This study investigates the roles of cyclooxygenase isoforms and thromboxane receptor activities in the differential endothelial dilatory capacities of arteries derived from omental and subcutaneous adipose tissues in obesity. MAIN METHODS Small arteries were isolated from omental and subcutaneous adipose tissues obtained from consented morbidly obese patients (n = 65, BMI 45 ± 6 kg m-2 [Mean ± SD]) undergoing bariatric surgery. Relaxation to acetylcholine was studied by wire myography in the absence or presence of indomethacin (10 μM, cyclooxygenase inhibitor), FR122047 (1 μM, cyclooxygenase-1 inhibitor), Celecoxib (4 μM, cyclooxygenase-2 inhibitor), Nω-Nitro-L-arginine methyl ester (L-NAME, 100 μM, nitric oxide synthase inhibitor) or combination of apamin (0.5 μM) and charybdotoxin (0.1 μM) that together inhibit endothelium-derived hyperpolarizing factor (EDHF). Contractions to U46619 (thromboxane A2 mimetic) were also studied. KEY FINDINGS Acetylcholine relaxation was significantly attenuated in omental compared with subcutaneous arteries from same patients (p < 0.01). Indomethacin (p < 0.01) and FR122047 (p < 0.001) but not Celecoxib significantly improved the omental arteriolar relaxation. Cyclooxygenase-1 mRNA and U46619 contractions were both increased in omental compared with subcutaneous arteries (p < 0.05). L-NAME comparably inhibited acetylcholine relaxation in both arteries, while apamin+charybdotoxin were less effective in omental compared with subcutaneous arteries. SIGNIFICANCE The results show that the depot-specific reduction in endothelial dilatory capacity of omental compared with subcutaneous arteries in obesity is in large part due to altered cyclooxygenase-1 and enhanced thromboxane receptor activities, which cause EDHF deficiency.
Collapse
Affiliation(s)
- Asmaa Raees
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | - Aysha Bakhamis
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | | | - Moataz Bashah
- Metabolic and Bariatric Surgery Department, Hamad Medical Corporation, Doha, Qatar
| | - Mashael Al-Jaber
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | - David Abraham
- Centre for Rheumatology and Connective Tissue Diseases, Division of Medicine, University College London, UK
| | - Lucie H Clapp
- Institute of Cardiovascular Sciences, University College London, UK
| | - Nelson N Orie
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar.
| |
Collapse
|
50
|
Riccardello GJ, Shastri DN, Changa AR, Thomas KG, Roman M, Prestigiacomo CJ, Gandhi CD. Influence of Relative Residence Time on Side-Wall Aneurysm Inception. Neurosurgery 2019; 83:574-581. [PMID: 28945849 DOI: 10.1093/neuros/nyx433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Relative residence time (RRT) is a marker of disturbed blood flow, marked by low magnitude and high oscillatory wall shear stress (WSS). The relation between solute residence time in proximity to the vascular endothelium and the atherosclerotic process is well appreciated in the literature. OBJECTIVE To assess the influence of RRT on side-wall aneurysm inception to better understand the role of atherosclerosis in aneurysm formation. METHODS Fourteen side-wall internal carotid artery aneurysms from the Aneurisk repository which met criteria for parent vessel reconstruction were reconstructed with Vascular Modeling Toolkit. Computational fluid dynamics analysis was carried out in Fluent. RRT was calculated in MATLAB (The MathWorks Inc, Natick, Massachusetts). We analyzed the results for correlations, defined as presence or absence of local elevations in RRT in specific regions of vasculature. RESULTS RRT was concluded to be negatively correlated with aneurysm inception in this study of side-wall internal carotid artery aneurysms, with 12/14 cases yielding the absence of local RRT elevations within or in close proximity of the removed ostium. Subsequent analysis of WSS showed that 11 of 14 aneurysms were formed in an atheroprotective environment, with only 1 of 14 formed in an atherogenic environment. Two models were found to be of indeterminate environment. CONCLUSION Atherogenesis and atherosclerosis have long been thought to be a major inciting factor responsible for the formation of aneurysms in the cerebral vasculature. We propose that inception of side-wall aneurysms occurs in hemodynamic environments that promote an atheroprotective endothelial phenotype and that the atheroprotective phenotype is therefore aneurysmogenic.
Collapse
Affiliation(s)
- Gerald J Riccardello
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Darshan N Shastri
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Abhinav R Changa
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Kiran G Thomas
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Max Roman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | | | - Chirag D Gandhi
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey.,Department of Radiology, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|