1
|
Mirza A, Hsu CPD, Rodriguez A, Alvarez P, Lou L, Sey M, Agarwal A, Ramaswamy S, Hutcheson J. Computational Model for Early-Stage Aortic Valve Calcification Shows Hemodynamic Biomarkers. Bioengineering (Basel) 2024; 11:955. [PMID: 39451331 PMCID: PMC11504039 DOI: 10.3390/bioengineering11100955] [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: 07/16/2024] [Revised: 08/13/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Heart disease is a leading cause of mortality, with calcific aortic valve disease (CAVD) being the most prevalent subset. Being able to predict this disease in its early stages is important for monitoring patients before they need aortic valve replacement surgery. Thus, this study explored hydrodynamic, mechanical, and hemodynamic differences in healthy and very mildly calcified porcine small intestinal submucosa (PSIS) bioscaffold valves to determine any notable parameters between groups that could, possibly, be used for disease tracking purposes. Three valve groups were tested: raw PSIS as a control and two calcified groups that were seeded with human valvular interstitial and endothelial cells (VICs/VECs) and cultivated in calcifying media. These two calcified groups were cultured in either static or bioreactor-induced oscillatory flow conditions. Hydrodynamic assessments showed metrics were below thresholds associated for even mild calcification. Young's modulus, however, was significantly higher in calcified valves when compared to raw PSIS, indicating the morphological changes to the tissue structure. Fluid-structure interaction (FSI) simulations agreed well with hydrodynamic results and, most notably, showed a significant increase in time-averaged wall shear stress (TAWSS) between raw and calcified groups. We conclude that tracking hemodynamics may be a viable biomarker for early-stage CAVD tracking.
Collapse
Affiliation(s)
- Asad Mirza
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| | - Chia-Pei Denise Hsu
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| | - Andres Rodriguez
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| | - Paulina Alvarez
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| | - Lihua Lou
- Department of Mechanical Engineering, Florida International University, Miami, FL 33174, USA; (L.L.); (M.S.); (A.A.)
| | - Matty Sey
- Department of Mechanical Engineering, Florida International University, Miami, FL 33174, USA; (L.L.); (M.S.); (A.A.)
| | - Arvind Agarwal
- Department of Mechanical Engineering, Florida International University, Miami, FL 33174, USA; (L.L.); (M.S.); (A.A.)
| | - Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| | - Joshua Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (C.-P.D.H.); (A.R.); (P.A.); (S.R.)
| |
Collapse
|
2
|
Fan L, Yao D, Fan Z, Zhang T, Shen Q, Tong F, Qian X, Xu L, Jiang C, Dong N. Beyond VICs: Shedding light on the overlooked VECs in calcific aortic valve disease. Biomed Pharmacother 2024; 178:117143. [PMID: 39024838 DOI: 10.1016/j.biopha.2024.117143] [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: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Calcific aortic valve disease (CAVD) is prevalent in developed nations and has emerged as a pressing global public health concern due to population aging. The precise etiology of this disease remains uncertain, and recent research has primarily focused on examining the role of valvular interstitial cells (VICs) in the development of CAVD. The predominant treatment options currently available involve open surgery and minimally invasive interventional surgery, with no efficacious pharmacological treatment. This article seeks to provide a comprehensive understanding of valvular endothelial cells (VECs) from the aspects of valvular endothelium-derived nitric oxide (NO), valvular endothelial mechanotransduction, valvular endothelial injury, valvular endothelial-mesenchymal transition (EndMT), and valvular neovascularization, which have received less attention, and aims to establish their role and interaction with VICs in CAVD. The ultimate goal is to provide new perspectives for the investigation of non-invasive treatment options for this disease.
Collapse
Affiliation(s)
- Lin Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingyi Yao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tailong Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Shen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqiang Tong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chen Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Yasin AT, Ali ET, Shari FH, Mohammed AN. Extensive study of CCN4, VCAM-1, MMP-3, and GM-CSF as reliable markers for disease activity in rheumatoid arthritis. J Taibah Univ Med Sci 2024; 19:885-900. [PMID: 39262670 PMCID: PMC11387248 DOI: 10.1016/j.jtumed.2024.08.001] [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: 03/15/2024] [Revised: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Background The involvement of Wnt-1-induced secreted protein-1 (WISP1/CCN4) in several inflammatory reaction has recently been proposed. Nevertheless, this protein's involvement in rheumatoid arthritis (RA) remains debated. Associations between poorly diagnosed RA and several classical markers derived from demography and biochemistry have been reported. Aim We sought to investigate the reliability and effectiveness of serum concentrations of CCN4, vascular cell adhesion molecule-1 (VCAM-1), matrix melloprotenase-3 (MMP-3), and granulocyte-macrophage colony-stimulating factor (GM-CSF) in monitoring and predicting RA and bone damage, and their correlation with RA disease course. Methods The study analyzed 128 patients with RA, comprising 68 newly diagnosed and 60 previously diagnosed patients, as well as 60 controls. Biomarker levels were measured with enzyme linked immuno-sorbent assays. Routine laboratory parameters such as serological, clinical, biochemical, and hematological parameters were additionally measured. Demography, anthropometry, and clinical symptom data were collected through interviews and a questionnaire. The joint disease activity score 28 (DAS28) was used to determine disease activity. Results Concentrations of four biomarkers were significantly higher in the RA group than the healthy controls. Elevated biomarker concentrations were also observed in patients with high, rather than moderate or low, DAS28-ESR activity status, except for monocyte count, hematocrit (%), and urea level. Furthermore, CCN4 level positively correlated with VCAM-1, MMP-3, and GM-CSF levels, DA-S28-CRP and DAS28-ESR. The levels of three predictive markers, CCN4, VCAM-1, and MMP-3, were elevated in non-treated patients, whereas GM-CSF level showed no difference. The highest area under the curve was 73.3% for CCN4, with 93.3% sensitivity and 64.7% specificity. Conclusion Our data suggest that CCN4 can be reliably used to indicate activity and therapeutic response associated with RA, thus facilitating earlier RA diagnosis.
Collapse
Affiliation(s)
- Ahmed T Yasin
- Basrah Hospital for Women and Children, Pharmacy Department, Basrah Health Directorate, Basrah, Iraq
| | - Eman T Ali
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Basrah, Iraq
| | - Falah H Shari
- College of Pharmacy, Clinical Laboratory Sciences Department, University of Basrah, Basrah, Iraq
| | - Ali N Mohammed
- Rheumatology Department, Alsayab Teaching Hospital, Basrah, Iraq
| |
Collapse
|
4
|
Luo H, Li L, Han S, Liu T. The role of monocyte/macrophage chemokines in pathogenesis of osteoarthritis: A review. Int J Immunogenet 2024; 51:130-142. [PMID: 38462560 DOI: 10.1111/iji.12664] [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: 12/02/2023] [Revised: 02/08/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024]
Abstract
Osteoarthritis (OA) is one of the most common degenerative diseases characterised by joint pain, swelling and decreased mobility, with its main pathological features being articular synovitis, cartilage degeneration and osteophyte formation. Inflammatory cytokines and chemokines secreted by activated immunocytes can trigger various inflammatory and immune responses in articular cartilage and synovium, contributing to the genesis and development of OA. A series of monocyte/macrophage chemokines, including monocyte chemotaxis protein (MCP)-1/CCL2, MCP2/CCL8, macrophage inflammatory protein (MIP)-1α/CCL3, MIP-1β/CCL4, MIP-3α/CCL20, regulated upon activation, normal T-cell expressed and secreted /CCL5, CCL17 and macrophage-derived chemokine/CCL22, was proven to transmit cell signals by binding to G protein-coupled receptors on recipient cell surface, mediating and promoting inflammation in OA joints. However, the underlying mechanism of these chemokines in the pathogenesis of OA remains still elusive. Here, published literature was reviewed, and the function and mechanisms of monocyte/macrophage chemokines in OA pathogenesis were summarised. The symptoms and disease progression of OA were found to be effectively alleviated when the expression of these chemokines is inhibited. Elucidating these mechanisms could contribute to further understand how OA develops and provide potential targets for the early diagnosis of arthritis and drug treatment to delay or even halt OA progression.
Collapse
Affiliation(s)
- Hao Luo
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Linfeng Li
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Song Han
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Liu
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
5
|
Yang C, Huang F, Fang H, Zang Y. Jiawei Shengjiangsan's Effect on Renal Injury in Diabetic Nephropathy Mice is Investigated via the PI3K/Akt/NF-κB Signaling Pathway. Diabetes Metab Syndr Obes 2024; 17:1687-1698. [PMID: 38629025 PMCID: PMC11020332 DOI: 10.2147/dmso.s456205] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Purpose This study aimed to investigate the intervention mechanism of Jiawei Shengjiangsan (JWSJS) on kidney injury in diabetic nephropathy mice. Methods Thirty 8-week-old db/db mice were randomly divided into five groups: model group, Perindopril group, and JWSJS low-, medium-, and high-dose groups (n=6 per group) based on body weight. Additionally, a blank control group was established consisting of 6 db/m mice aged 8 weeks. The blank and model groups received daily intragastric administration of 7g/kg/d pure water. The remaining groups were assigned to JWSJS low (3.5g/kg/d), medium (7g/kg/d), high (14g/kg/d) dosage groups, and perindopril positive control group (0.48mg/kg/d) for 12 weeks. Post-experiment, serum creatinine (SCr) and blood urea nitrogen (BUN) were analyzed using an automatic biochemical analyzer. Enzyme-linked immunosorbent assay (ELISA) measured 24-hour urinary albumin, neutrophil gelatinase-associated lipocalin (NGAL), TNF-α, IL-1β, VCAM-1, MCP-1, and HbA1c. Western blot assessed the protein expressions of p-PI3K, p-Akt, and p-NF-κB p65, while pathological kidney changes were observed. Results Compared to the blank group, the model group exhibited increased SCr, BUN, 24-hour urinary albumin, serum NGAL, TNF-α, IL-1β, VCAM-1, MCP-1, HbA1c, p-PI3K, and p-Akt, alongside increased p-NF-κB p65 expression, indicating significant kidney pathology. After treatment, the JWSJS group showed decreased SCr, BUN, 24-hour urinary microalbumin, NGAL, HbA1c, TNF-α, IL-1β, VCAM-1, MCP-1 levels, increased p-PI3K and p-Akt expression (P<0.05), and reduced p-NF-κB p65 content (P<0.05). Histopathological analysis revealed that JWSJS ameliorated renal tubular epithelial cell damage, glomerular capillary and basement membrane injuries, and facilitated the repair of damaged podocytes in diabetic nephropathy mice. Conclusion JWSJS demonstrated efficacy in reducing renal inflammation in diabetic nephropathy mice, with its mechanism likely associated with the inhibition of the PI3K/Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chenhua Yang
- General Medicine, Bao’an Authentic TCM Therapy Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Fengling Huang
- College of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People’s Republic of China
| | - Huiqin Fang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yunhua Zang
- General Medicine, Bao’an Authentic TCM Therapy Hospital, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
6
|
Azimi-Boulali J, Mahler GJ, Murray BT, Huang P. Multiscale computational modeling of aortic valve calcification. Biomech Model Mechanobiol 2024; 23:581-599. [PMID: 38093148 DOI: 10.1007/s10237-023-01793-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/13/2023] [Indexed: 03/26/2024]
Abstract
Calcific aortic valve disease (CAVD) is a common cardiovascular disease that affects millions of people worldwide. The disease is characterized by the formation of calcium nodules on the aortic valve leaflets, which can lead to stenosis and heart failure if left untreated. The pathogenesis of CAVD is still not well understood, but involves several signaling pathways, including the transforming growth factor beta (TGF β ) pathway. In this study, we developed a multiscale computational model for TGF β -stimulated CAVD. The model framework comprises cellular behavior dynamics, subcellular signaling pathways, and tissue-level diffusion fields of pertinent chemical species, where information is shared among different scales. Processes such as endothelial to mesenchymal transition (EndMT), fibrosis, and calcification are incorporated. The results indicate that the majority of myofibroblasts and osteoblast-like cells ultimately die due to lack of nutrients as they become trapped in areas with higher levels of fibrosis or calcification, and they subsequently act as sources for calcium nodules, which contribute to a polydispersed nodule size distribution. Additionally, fibrosis and calcification processes occur more frequently in regions closer to the endothelial layer where the cell activity is higher. Our results provide insights into the mechanisms of CAVD and TGF β signaling and could aid in the development of novel therapeutic approaches for CAVD and other related diseases such as cancer. More broadly, this type of modeling framework can pave the way for unraveling the complexity of biological systems by incorporating several signaling pathways in subcellular models to simulate tissue remodeling in diseases involving cellular mechanobiology.
Collapse
Affiliation(s)
- Javid Azimi-Boulali
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Bruce T Murray
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Peter Huang
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA.
| |
Collapse
|
7
|
Vogl B, Sularz A, Lilly S, Thourani VH, Lindman BR, Alkhouli M, Hatoum H. Effect of Blood Pressure Levels on Sinus Hemodynamics in Relation to Calcification After Bioprosthetic Aortic Valve Replacement. Ann Biomed Eng 2024; 52:888-897. [PMID: 38157105 DOI: 10.1007/s10439-023-03426-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Coexisting hypertension and aortic stenosis are common. Some studies showed that elevated blood pressures may be associated with progression of calcific aortic valve disease (CAVD) while others showed no correlation. Flow dynamics in the sinuses of Valsalva are considered key factors in the progression of CAVD. While the relationship between hemodynamics and CAVD is not yet fully understood, it has been demonstrated that they are tightly correlated. This study aims to investigate the effect of changing systolic and diastolic blood pressures (SBP and DBP, respectively) on sinus hemodynamics in relation to potential initiation or progression of CAVD after aortic valve replacement (AVR). Evolut R, SAPIEN 3 and Magna valves were deployed in an aortic root under pulsatile conditions. Using particle image velocimetry, the hemodynamics in the sinus were assessed. The velocity, vorticity, circulation ( Γ ) and shear stress were calculated. This study shows that under elevated SBP and DBP, velocity, vorticity, and shear stress nearby the leaflets increased. Additionally, larger fluctuations of Γ and area under the curve throughout the cardiac cycle were observed. Elevated blood pressures are associated with higher velocity, vorticity, and shear stress near the leaflets which may initiate or accelerate pro-calcific changes in the prosthetic leaflets leading to bioprosthetic valve degeneration.
Collapse
Affiliation(s)
- Brennan Vogl
- Biomedical Engineering Department, Michigan Technological University, 1400 Townsend Dr, Houghton, MI, 49931, USA
| | - Agata Sularz
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Scott Lilly
- Department of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Vinod H Thourani
- Department of Cardiovascular Surgery, Marcus Valve Center, Piedmont Heart Institute, Atlanta, GA, USA
| | - Brian R Lindman
- Division of Cardiovascular Medicine, Structural Heart and Valve Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohamad Alkhouli
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hoda Hatoum
- Biomedical Engineering Department, Michigan Technological University, 1400 Townsend Dr, Houghton, MI, 49931, USA.
- Health Research Institute, Center of Biocomputing and Digital Health and Institute of Computing and Cybernetics, Michigan Technological University, Houghton, MI, USA.
| |
Collapse
|
8
|
Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 2024; 25:390-406. [PMID: 35751379 PMCID: PMC9974241 DOI: 10.1177/11297298221085458] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Loay H Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
9
|
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
|
10
|
Dittfeld C, Winkelkotte M, Scheer A, Voigt E, Schmieder F, Behrens S, Jannasch A, Matschke K, Sonntag F, Tugtekin SM. Challenges of aortic valve tissue culture - maintenance of viability and extracellular matrix in the pulsatile dynamic microphysiological system. J Biol Eng 2023; 17:60. [PMID: 37770970 PMCID: PMC10538250 DOI: 10.1186/s13036-023-00377-1] [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: 05/15/2023] [Accepted: 09/14/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) causes an increasing health burden in the 21st century due to aging population. The complex pathophysiology remains to be understood to develop novel prevention and treatment strategies. Microphysiological systems (MPSs), also known as organ-on-chip or lab-on-a-chip systems, proved promising in bridging in vitro and in vivo approaches by applying integer AV tissue and modelling biomechanical microenvironment. This study introduces a novel MPS comprising different micropumps in conjunction with a tissue-incubation-chamber (TIC) for long-term porcine and human AV incubation (pAV, hAV). RESULTS Tissue cultures in two different MPS setups were compared and validated by a bimodal viability analysis and extracellular matrix transformation assessment. The MPS-TIC conjunction proved applicable for incubation periods of 14-26 days. An increased metabolic rate was detected for pulsatile dynamic MPS culture compared to static condition indicated by increased LDH intensity. ECM changes such as an increase of collagen fibre content in line with tissue contraction and mass reduction, also observed in early CAVD, were detected in MPS-TIC culture, as well as an increase of collagen fibre content. Glycosaminoglycans remained stable, no significant alterations of α-SMA or CD31 epitopes and no accumulation of calciumhydroxyapatite were observed after 14 days of incubation. CONCLUSIONS The presented ex vivo MPS allows long-term AV tissue incubation and will be adopted for future investigation of CAVD pathophysiology, also implementing human tissues. The bimodal viability assessment and ECM analyses approve reliability of ex vivo CAVD investigation and comparability of parallel tissue segments with different treatment strategies regarding the AV (patho)physiology.
Collapse
Affiliation(s)
- Claudia Dittfeld
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany.
| | - Maximilian Winkelkotte
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Anna Scheer
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Emmely Voigt
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Florian Schmieder
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Stephan Behrens
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Anett Jannasch
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Klaus Matschke
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| | - Frank Sonntag
- Fraunhofer Institute for Material and Beam Technology IWS, Dresden, Germany
| | - Sems-Malte Tugtekin
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstr. 76, 01307, Dresden, Germany
| |
Collapse
|
11
|
Lee W, Jung K, Song H, Lee H, Park HE, Koh Y, Choi S, Park K. Clonal hematopoiesis with DNMT3A mutation is associated with lower white matter hyperintensity volume. CNS Neurosci Ther 2023; 29:1243-1253. [PMID: 36807865 PMCID: PMC10068463 DOI: 10.1111/cns.14114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/22/2022] [Accepted: 01/20/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) increases the risk of cerebrovascular events, while its association with cerebral white matter hyperintensity (WMH) is undemonstrated. We evaluated the effect of CHIP and its major driving mutations on cerebral WMH severity. METHODS From an institutional cohort of a routine health check-up program with a DNA repository database, subjects who were ≥50 years of age, with one or more cardiovascular risk factors but no central nervous system disorder, and performed brain MRI were included. Along with the presence of CHIP and its major driving mutations, clinical and laboratory data were obtained. WMH volume was measured in total, periventricular, and subcortical regions. RESULTS Among the total 964 subjects, 160 subjects were classified as CHIP positive group. CHIP was most frequently associated with DNMT3A mutation (48.8%), followed by TET2 (11.9%) and ASXL1 (8.1%) mutations. Linear regression analysis adjusting for age, sex, and conventional cerebrovascular risk factors suggested that CHIP with DNMT3A mutation was associated with the lower log-transformed total WMH volume, unlike other CHIP mutations. When classified according to variant allele fraction (VAF) value of DNMT3A mutation, higher VAF classes were associated with the lower log-transformed total WMH and the lower log-transformed periventricular WMH volume, but not with the log-transformed subcortical WMH volumes. CONCLUSIONS Clonal hematopoiesis with DNMT3A mutation is quantitatively associated with a lower volume of cerebral WMH, especially in the periventricular region. CHIP with DNMT3A mutation might have a protective role in the endothelial pathomechanism of WMH.
Collapse
Affiliation(s)
- Woo‐Jin Lee
- Department of NeurologySeoul National University Bundang HospitalSeongnam‐siSouth Korea
- Department of NeurologySeoul National University HospitalSeoulSouth Korea
| | - Keun‐Hwa Jung
- Department of NeurologySeoul National University HospitalSeoulSouth Korea
| | - Han Song
- Genome Opinion Inc.SeoulSouth Korea
| | - Heesun Lee
- Division of Cardiology, Department of Internal MedicineSeoul National University Healthcare System Gangnam CenterSeoulSouth Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulSouth Korea
| | - Hyo Eun Park
- Division of Cardiology, Department of Internal MedicineSeoul National University Healthcare System Gangnam CenterSeoulSouth Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulSouth Korea
| | - Youngil Koh
- Genome Opinion Inc.SeoulSouth Korea
- Division of Hemato‐oncology, Department of Internal MedicineSeoul National University HospitalSeoulSouth Korea
| | - Su‐Yeon Choi
- Division of Cardiology, Department of Internal MedicineSeoul National University Healthcare System Gangnam CenterSeoulSouth Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulSouth Korea
| | - Kyung‐Il Park
- Department of NeurologySeoul National University HospitalSeoulSouth Korea
- Department of NeurologySeoul National University Healthcare System Gangnam CenterSeoulSouth Korea
| |
Collapse
|
12
|
Shu L, Yuan Z, Li F, Cai Z. Oxidative stress and valvular endothelial cells in aortic valve calcification. Biomed Pharmacother 2023; 163:114775. [PMID: 37116353 DOI: 10.1016/j.biopha.2023.114775] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/15/2023] [Accepted: 04/23/2023] [Indexed: 04/30/2023] Open
Abstract
Calcified aortic valve disease (CAVD) is a common cardiovascular disease in elderly individuals. Although it was previously considered a degenerative disease, it is, in fact, a progressive disease involving multiple mechanisms. Aortic valve endothelial cells, which cover the outermost layer of the aortic valve and are directly exposed to various pathogenic factors, play a significant role in the onset and progression of CAVD. Hemodynamic changes can directly damage the structure and function of valvular endothelial cells (VECs). This leads to inflammatory infiltration and oxidative stress, which promote the progression of CAVD. VECs can regulate the pathological differentiation of valvular interstitial cells (VICs) through NO and thus affect the process of CAVD. Under the influence of pathological factors, VECs can also be transformed into VICs through EndMT, and then the pathological differentiation of VICs eventually leads to the formation of calcification. This review discusses the role of VECs, especially the role of oxidative stress in VECs, in the process of aortic valve calcification.
Collapse
Affiliation(s)
- Li Shu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Zhen Yuan
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Zhejun Cai
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
13
|
Vinton Z, Wolfe K, Fisher J, Brooks A. The Effect of Celecoxib on the Progression of Calcific Aortic Valve Disease-Protective or Pathogenic? J Clin Med 2023; 12:jcm12072717. [PMID: 37048799 PMCID: PMC10094907 DOI: 10.3390/jcm12072717] [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: 02/17/2023] [Revised: 03/24/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is a debilitating condition for which there are limited therapeutic options aside from valve replacement. As such, it is crucial to explore alternative management strategies for CAVD. Non-steroidal anti-inflammatory drugs (NSAIDs), particularly celecoxib, have been the subject of debate in the literature regarding their potential impact on CAVD. We conducted an in-depth analysis of five studies exploring the effect of celecoxib on CAVD and found discrepancies in both methods and results. Our findings suggest that celecoxib may impact the development of this disease via multiple mechanisms, each of which may have different effects on its pathogenesis. We also discovered limited clinical research examining the connection between celecoxib use and CAVD in medical patients. As such, further studies are needed to clarify the role of celecoxib and other NSAIDs in CAVD progression in order to inform future treatment options and clarify their impact on the disease.
Collapse
Affiliation(s)
- Zachary Vinton
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80112, USA
| | - Kevin Wolfe
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80112, USA
| | - Jensen Fisher
- Department of Library Services, Rocky Vista University, Parker, CO 80112, USA
| | - Amanda Brooks
- Office of Research and Scholarly Activity, Rocky Vista University, Parker, CO 80112, USA
| |
Collapse
|
14
|
Abi Rached NM, Gbotosho OT, Archer DR, Jones JA, Sterling MS, Hyacinth HI. Adhesion molecules and cerebral microvascular hemodynamic abnormalities in sickle cell disease. Front Neurol 2022; 13:976063. [PMID: 36570439 PMCID: PMC9767957 DOI: 10.3389/fneur.2022.976063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebrovascular abnormalities are a common feature of sickle cell disease that may be associated with risk of vaso-occlusive pain crises, microinfarcts, and cognitive impairment. An activated endothelium and adhesion factors, VCAM-1 and P-selectin, are implicated in sickle cell vasculopathy, including abnormal hemodynamics and leukocyte adherence. This study examined the association between cerebral expression of these adhesion factors and cortical microvascular blood flow dynamics by using in-vivo two-photon microscopy. We also examined the impact of blood transfusion treatment on these markers of vasculopathy. Results showed that sickle cell mice had significantly higher maximum red blood cell (RBC) velocity (6.80 ± 0.25 mm/sec, p ≤ 0.01 vs. 5.35 ± 0.35 mm/sec) and more frequent blood flow reversals (18.04 ± 0.95 /min, p ≤ 0.01 vs. 13.59 ± 1.40 /min) in the cortical microvasculature compared to controls. In addition, sickle cell mice had a 2.6-fold (RFU/mm2) increase in expression of VCAM-1 and 17-fold (RFU/mm2) increase in expression of P-selectin compared to controls. This was accompanied by an increased frequency in leukocyte adherence (4.83 ± 0.57 /100 μm/min vs. 2.26 ± 0.37 /100 μm/min, p ≤ 0.001). We also found that microinfarcts identified in sickle cell mice were 50% larger than in controls. After blood transfusion, many of these parameters improved, as results demonstrated that sickle cell mice had a lower post-transfusion maximum RBC velocity (8.30 ± 0.98 mm/sec vs. 11.29 ± 0.95 mm/sec), lower frequency of blood flow reversals (12.80 ± 2.76 /min vs. 27.75 ± 2.09 /min), and fewer instances of leukocyte adherence compared to their pre-transfusion imaging time point (1.35 ± 0.32 /100 μm/min vs. 3.46 ± 0.58 /100 μm/min). Additionally, we found that blood transfusion was associated with lower expression of adhesion factors. Our results suggest that blood transfusion and adhesion factors, VCAM-1 and P-selectin, are potential therapeutic targets for addressing cerebrovascular pathology, such as vaso-occlusion, in sickle cell disease.
Collapse
Affiliation(s)
- Noor Mary Abi Rached
- Neuroscience and Behavioral Biology Undergraduate Program, Emory University, Atlanta, GA, United States
| | - Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David R. Archer
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jayre A. Jones
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Morgan S. Sterling
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
15
|
Salim MT, Villa-Roel N, Vogel B, Jo H, Yoganathan AP. HIF1A inhibitor PX-478 reduces pathological stretch-induced calcification and collagen turnover in aortic valve. Front Cardiovasc Med 2022; 9:1002067. [PMID: 36419483 PMCID: PMC9676244 DOI: 10.3389/fcvm.2022.1002067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/20/2022] [Indexed: 09/19/2023] Open
Abstract
HIF1A is significantly upregulated in calcified human aortic valves (AVs). Furthermore, HIF1A inhibitor PX-478 was shown to inhibit AV calcification under static and disturbed flow conditions. Since elevated stretch is one of the major mechanical stimuli for AV calcification, we investigated the effect of PX-478 on AV calcification and collagen turnover under a pathophysiological cyclic stretch (15%) condition. Porcine aortic valve (PAV) leaflets were cyclically (1 Hz) stretched at 15% for 24 days in osteogenic medium with or without PX-478. In addition, PAV leaflets were cyclically stretched at a physiological (10%) and 15% for 3 days in regular medium to assess its effect of on HIF1A mRNA expression. It was found that 100 μM (high concentration) PX-478 could significantly inhibit PAV calcification under 15% stretch, whereas 50 μM (moderate concentration) PX-478 showed a modest inhibitory effect on PAV calcification. Nonetheless, 50 μM PX-478 significantly reduced PAV collagen turnover under 15% stretch. Surprisingly, it was observed that cyclic stretch (15% vs. 10%) did not have any significant effect on HIF1A mRNA expression in PAV leaflets. These results suggest that HIF1A inhibitor PX-478 may impart its anti-calcific and anti-matrix remodeling effect in a stretch-independent manner.
Collapse
Affiliation(s)
- Md Tausif Salim
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Nicolas Villa-Roel
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Booth Vogel
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| | - Hanjoong Jo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Ajit P. Yoganathan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| |
Collapse
|
16
|
Chen J, Liu Q, He J, Li Y. Immune responses in diabetic nephropathy: Pathogenic mechanisms and therapeutic target. Front Immunol 2022; 13:958790. [PMID: 36045667 PMCID: PMC9420855 DOI: 10.3389/fimmu.2022.958790] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/28/2022] [Indexed: 11/14/2022] Open
Abstract
Diabetic nephropathy (DN) is a chronic, inflammatory disease affecting millions of diabetic patients worldwide. DN is associated with proteinuria and progressive slowing of glomerular filtration, which often leads to end-stage kidney diseases. Due to the complexity of this metabolic disorder and lack of clarity about its pathogenesis, it is often more difficult to diagnose and treat than other kidney diseases. Recent studies have highlighted that the immune system can inadvertently contribute to DN pathogenesis. Cells involved in innate and adaptive immune responses can target the kidney due to increased expression of immune-related localization factors. Immune cells then activate a pro-inflammatory response involving the release of autocrine and paracrine factors, which further amplify inflammation and damage the kidney. Consequently, strategies to treat DN by targeting the immune responses are currently under study. In light of the steady rise in DN incidence, this timely review summarizes the latest findings about the role of the immune system in the pathogenesis of DN and discusses promising preclinical and clinical therapies.
Collapse
Affiliation(s)
| | | | - Jinhan He
- *Correspondence: Jinhan He, ; Yanping Li,
| | - Yanping Li
- *Correspondence: Jinhan He, ; Yanping Li,
| |
Collapse
|
17
|
Zhou Y, Shi W, Zhao D, Xiao S, Wang K, Wang J. Identification of Immune-Associated Genes in Diagnosing Aortic Valve Calcification With Metabolic Syndrome by Integrated Bioinformatics Analysis and Machine Learning. Front Immunol 2022; 13:937886. [PMID: 35865542 PMCID: PMC9295723 DOI: 10.3389/fimmu.2022.937886] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022] Open
Abstract
Background Immune system dysregulation plays a critical role in aortic valve calcification (AVC) and metabolic syndrome (MS) pathogenesis. The study aimed to identify pivotal diagnostic candidate genes for AVC patients with MS. Methods We obtained three AVC and one MS dataset from the gene expression omnibus (GEO) database. Identification of differentially expressed genes (DEGs) and module gene via Limma and weighted gene co-expression network analysis (WGCNA), functional enrichment analysis, protein–protein interaction (PPI) network construction, and machine learning algorithms (least absolute shrinkage and selection operator (LASSO) regression and random forest) were used to identify candidate immune-associated hub genes for diagnosing AVC with MS. To assess the diagnostic value, the nomogram and receiver operating characteristic (ROC) curve were developed. Finally, immune cell infiltration was created to investigate immune cell dysregulation in AVC. Results The merged AVC dataset included 587 DEGs, and 1,438 module genes were screened out in MS. MS DEGs were primarily enriched in immune regulation. The intersection of DEGs for AVC and module genes for MS was 50, which were mainly enriched in the immune system as well. Following the development of the PPI network, 26 node genes were filtered, and five candidate hub genes were chosen for nomogram building and diagnostic value evaluation after machine learning. The nomogram and all five candidate hub genes had high diagnostic values (area under the curve from 0.732 to 0.982). Various dysregulated immune cells were observed as well. Conclusion Five immune-associated candidate hub genes (BEX2, SPRY2, CXCL16, ITGAL, and MORF4L2) were identified, and the nomogram was constructed for AVC with MS diagnosis. Our study could provide potential peripheral blood diagnostic candidate genes for AVC in MS patients.
Collapse
Affiliation(s)
- Yufei Zhou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenxiang Shi
- Department of Pediatric Cardiology, Xinhua Hospital, The Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Zhao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Kai Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jing Wang, ; Kai Wang,
| | - Jing Wang
- Department of Geriatric Medicine, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
- *Correspondence: Jing Wang, ; Kai Wang,
| |
Collapse
|
18
|
He L, Zhang CL, Chen Q, Wang L, Huang Y. Endothelial shear stress signal transduction and atherogenesis: From mechanisms to therapeutics. Pharmacol Ther 2022; 235:108152. [PMID: 35122834 DOI: 10.1016/j.pharmthera.2022.108152] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/13/2022] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Atherosclerotic vascular disease and its complications are among the top causes of mortality worldwide. In the vascular lumen, atherosclerotic plaques are not randomly distributed. Instead, they are preferentially localized at the curvature and bifurcations along the arterial tree, where shear stress is low or disturbed. Numerous studies demonstrate that endothelial cell phenotypic change (e.g., inflammation, oxidative stress, endoplasmic reticulum stress, apoptosis, autophagy, endothelial-mesenchymal transition, endothelial permeability, epigenetic regulation, and endothelial metabolic adaptation) induced by oscillatory shear force play a fundamental role in the initiation and progression of atherosclerosis. Mechano-sensors, adaptor proteins, kinases, and transcriptional factors work closely at different layers to transduce the shear stress force from the plasma membrane to the nucleus in endothelial cells, thereby controlling the expression of genes that determine cell fate and phenotype. An in-depth understanding of these mechano-sensitive signaling cascades shall provide new translational strategies for therapeutic intervention of atherosclerotic vascular disease. This review updates the recent advances in endothelial mechano-transduction and its role in the pathogenesis of atherosclerosis, and highlights the perspective of new anti-atherosclerosis therapies through targeting these mechano-regulated signaling molecules.
Collapse
Affiliation(s)
- Lei He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Qinghua Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Oxidative Stress in Calcific Aortic Valve Stenosis: Protective Role of Natural Antioxidants. Antioxidants (Basel) 2022; 11:antiox11061169. [PMID: 35740065 PMCID: PMC9219756 DOI: 10.3390/antiox11061169] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 01/01/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is the most prevalent heart valvular disease worldwide and a slowly progressive disorder characterized by thickening of the aortic valve, calcification, and subsequent heart failure. Valvular calcification is an active cell regulation process in which valvular interstitial cells involve phenotypic conversion into osteoblasts/chondrocytes-like cells. The underlying pathophysiology is complicated, and there have been no pharmacological treatments for CAVS to date. Recent studies have suggested that an increase in oxidative stress is the major trigger of CAVS, and natural antioxidants could ameliorate the detrimental effects of reactive oxygen species in the pathogenesis of CAVS. It is imperative to review the current findings regarding the role of natural antioxidants in CAVS, as they can be a promising therapeutic approach for managing CAVS, a disorder currently without effective treatment. This review summarizes the current findings on molecular mechanisms associated with oxidative stress in the development of valvular calcification and discusses the protective roles of natural antioxidants in the prevention and treatment of CAVS.
Collapse
|
20
|
The Haemodynamic and Pathophysiological Mechanisms of Calcific Aortic Valve Disease. Biomedicines 2022; 10:biomedicines10061317. [PMID: 35740339 PMCID: PMC9220142 DOI: 10.3390/biomedicines10061317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
|
21
|
Han K, Xia Y, Shi D, Yang L, Xie M, Wang Z, Gao F, Shao Q, Ma X, Zhou Y. Relation of Monocyte Number to Progression of Aortic Stenosis. Am J Cardiol 2022; 171:122-126. [PMID: 35341577 DOI: 10.1016/j.amjcard.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 11/15/2022]
Abstract
Rapid progression of aortic stenosis (AS) is associated with poor prognosis. However, the relation between monocyte number and AS progression is unknown. Here, we detected the relation between monocyte number and AS progression. We retrospectively analyzed 220 patients with AS with at least 2 echocardiograms with the maximal interval ≥180 days from January 2016 to June 2021. AS severity was categorized by aortic jet velocity (Vmax) and mean pressure gradient. Rapid progression of AS was defined when Vmax increased ≥0.3 m/s/year. Patients were divided into low and high monocyte groups according to the cut-off value of the receiver-operating characteristic curve. AS progression was compared between the 2 groups. Various models of binary logistic regression were used to reveal the association between monocyte number and rapid progression. During a median of 601 days of echocardiographic follow-up (interquartile range 353 to 909), 52.7% of the population was in rapid progression. Patients in the high monocyte group had more rapid progression in both Vmax and mean pressure gradient (p = 0.020 and p = 0.030, respectively). The percentage of patients with severe AS was increased by 5.4% in the low monocyte group and 16.9% in the high monocyte group. Different models of binary logistic regression showed that the monocyte number was positively associated with the rapid progression. In conclusion, a higher monocyte number was associated with the rapid progression of AS.
Collapse
Affiliation(s)
- Kangning Han
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Yihua Xia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Dongmei Shi
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Lixia Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Meng Xie
- Department of Echocardiogram, Beijing Anzhen Hospital, Capital Medical University
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Fei Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Qiaoyu Shao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Xiaoteng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University..
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University..
| |
Collapse
|
22
|
Tyagi N, Gambhir K, Sharma D, Gangenahalli G, Verma YK. Data mining and structural analysis for multi-tissue regeneration potential of BMP-4 and activator drugs. J Biomol Struct Dyn 2022:1-16. [DOI: 10.1080/07391102.2022.2067899] [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]
Affiliation(s)
- Nishant Tyagi
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Delhi, India
| | - Kirtida Gambhir
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Delhi, India
| | - Deepak Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Gurudutta Gangenahalli
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Delhi, India
| | - Yogesh Kumar Verma
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organisation (DRDO), Delhi, India
| |
Collapse
|
23
|
Mendoza M, Chen MH, Huang P, Mahler GJ. Shear and endothelial induced late-stage calcific aortic valve disease-on-a-chip develops calcium phosphate mineralizations. LAB ON A CHIP 2022; 22:1374-1385. [PMID: 35234762 DOI: 10.1039/d1lc00931a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Calcific aortic valve disease (CAVD) is an active pathobiological process leading to severe aortic stenosis, where the only treatment is valve replacement. Late-stage CAVD is characterized by calcification, disorganization of collagen, and deposition of glycosaminoglycans, such as chondroitin sulfate (CS), in the fibrosa. We developed a three-dimensional microfluidic device of the aortic valve fibrosa to study the effects of shear stress (1 or 20 dyne per cm2), CS (1 or 20 mg mL-1), and endothelial cell presence on calcification. CAVD chips consisted of a collagen I hydrogel, where porcine aortic valve interstitial cells were embedded within and porcine aortic valve endothelial cells were seeded on top of the matrix for up to 21 days. Here, we show that this CAVD-on-a-chip is the first to develop human-like calcified nodules varying in calcium phosphate mineralization maturity resulting from high shear and endothelial cells, specifically di- and octa-calcium phosphates. Long-term co-culture microfluidic studies confirmed cell viability and calcium phosphate formations throughout 21 days. Given that CAVD has no targeted therapies, the creation of a physiologically relevant test-bed of the aortic valve could lead to advances in preclinical studies.
Collapse
Affiliation(s)
- Melissa Mendoza
- Department of Biomedical Engineering, Binghamton University, P.O Box 6000, Binghamton, NY, 13902, USA.
| | - Mei-Hsiu Chen
- Department of Mathematical Sciences, Binghamton University, Binghamton, NY, 13902, USA
| | - Peter Huang
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, P.O Box 6000, Binghamton, NY, 13902, USA.
| |
Collapse
|
24
|
Dayawansa NH, Baratchi S, Peter K. Uncoupling the Vicious Cycle of Mechanical Stress and Inflammation in Calcific Aortic Valve Disease. Front Cardiovasc Med 2022; 9:783543. [PMID: 35355968 PMCID: PMC8959593 DOI: 10.3389/fcvm.2022.783543] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a common acquired valvulopathy, which carries a high burden of mortality. Chronic inflammation has been postulated as the predominant pathophysiological process underlying CAVD. So far, no effective medical therapies exist to halt the progression of CAVD. This review aims to outline the known pathways of inflammation and calcification in CAVD, focussing on the critical roles of mechanical stress and mechanosensing in the perpetuation of valvular inflammation. Following initiation of valvular inflammation, dysregulation of proinflammatory and osteoregulatory signalling pathways stimulates endothelial-mesenchymal transition of valvular endothelial cells (VECs) and differentiation of valvular interstitial cells (VICs) into active myofibroblastic and osteoblastic phenotypes, which in turn mediate valvular extracellular matrix remodelling and calcification. Mechanosensitive signalling pathways convert mechanical forces experienced by valve leaflets and circulating cells into biochemical signals and may provide the positive feedback loop that promotes acceleration of disease progression in the advanced stages of CAVD. Mechanosensing is implicated in multiple aspects of CAVD pathophysiology. The mechanosensitive RhoA/ROCK and YAP/TAZ systems are implicated in aortic valve leaflet mineralisation in response to increased substrate stiffness. Exposure of aortic valve leaflets, endothelial cells and platelets to high shear stress results in increased expression of mediators of VIC differentiation. Upregulation of the Piezo1 mechanoreceptor has been demonstrated to promote inflammation in CAVD, which normalises following transcatheter valve replacement. Genetic variants and inhibition of Notch signalling accentuate VIC responses to altered mechanical stresses. The study of mechanosensing pathways has revealed promising insights into the mechanisms that perpetuate inflammation and calcification in CAVD. Mechanotransduction of altered mechanical stresses may provide the sought-after coupling link that drives a vicious cycle of chronic inflammation in CAVD. Mechanosensing pathways may yield promising targets for therapeutic interventions and prognostic biomarkers with the potential to improve the management of CAVD.
Collapse
Affiliation(s)
- Nalin H. Dayawansa
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Sara Baratchi
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Pham DH, Dai CR, Lin B, Butcher JT. Local fluid shear stress operates a molecular switch to drive fetal semilunar valve extension. Dev Dyn 2022; 251:481-497. [PMID: 34535945 PMCID: PMC8891031 DOI: 10.1002/dvdy.419] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND While much is known about the genetic regulation of early valvular morphogenesis, mechanisms governing fetal valvular growth and remodeling remain unclear. Hemodynamic forces strongly influence morphogenesis, but it is unknown whether or how they interact with valvulogenic signaling programs. Side-specific activity of valvulogenic programs motivates the hypothesis that shear stress pattern-specific endocardial signaling controls the elongation of leaflets. RESULTS We determined that extension of the semilunar valve occurs via fibrosa sided endocardial proliferation. Low OSS was necessary and sufficient to induce canonical Wnt/β-catenin activation in fetal valve endothelium, which in turn drives BMP receptor/ligand expression, and pSmad1/5 activity essential for endocardial proliferation. In contrast, ventricularis endocardial cells expressed active Notch1 but minimal pSmad1/5. Endocardial monolayers exposed to LSS attenuate Wnt signaling in a Notch1 dependent manner. CONCLUSIONS Low OSS is transduced by endocardial cells into canonical Wnt signaling programs that regulate BMP signaling and endocardial proliferation. In contrast, high LSS induces Notch signaling in endocardial cells, inhibiting Wnt signaling and thereby restricting growth on the ventricular surface. Our results identify a novel mechanically regulated molecular switch, whereby fluid shear stress drives the growth of valve endothelium, orchestrating the extension of the valve in the direction of blood flow.
Collapse
Affiliation(s)
- Duc H. Pham
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Charles R. Dai
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Belle Lin
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jonathan T. Butcher
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Corresponding author:
| |
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: 3] [Impact Index Per Article: 1.5] [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
|
Innate immune cells in the pathophysiology of calcific aortic valve disease: lessons to be learned from atherosclerotic cardiovascular disease? Basic Res Cardiol 2022; 117:28. [PMID: 35581364 PMCID: PMC9114076 DOI: 10.1007/s00395-022-00935-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 01/31/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular disease in the developed world with currently no effective pharmacological treatment available. CAVD results from a complex, multifactorial process, in which valvular inflammation and fibro-calcific remodelling lead to valve thickening and cardiac outflow obstruction. The exact underlying pathophysiology of CAVD is still not fully understood, yet the development of CAVD shows many similarities with the pathophysiology of atherosclerotic cardiovascular disease (ASCVD), such as coronary artery disease. Innate immune cells play a crucial role in ASCVD and might also play a pivotal role in the development of CAVD. This review summarizes the current knowledge on the role of innate immune cells, both in the circulation and in the aortic valve, in the development of CAVD and the similarities and differences with ASCVD. Trained immunity and clonal haematopoiesis of indeterminate potential are proposed as novel immunological mechanisms that possibly contribute to the pathophysiology of CAVD and new possible treatment targets are discussed.
Collapse
|
28
|
Deb N, Lacerda CMR. Valvular Endothelial Cell Response to the Mechanical Environment-A Review. Cell Biochem Biophys 2021; 79:695-709. [PMID: 34661855 DOI: 10.1007/s12013-021-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Heart valve leaflets are complex structures containing valve endothelial cells, interstitial cells, and extracellular matrix. Heart valve endothelial cells sense mechanical stimuli, and communicate amongst themselves and the surrounding cells and extracellular matrix to maintain tissue homeostasis. In the presence of abnormal mechanical stimuli, endothelial cell communication is triggered in defense and such processes may eventually lead to cardiac disease progression. This review focuses on the role of mechanical stimuli on heart valve endothelial surfaces-from heart valve development and maintenance of tissue integrity to disease progression with related signal pathways involved in this process.
Collapse
Affiliation(s)
- Nandini Deb
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US
| | - Carla M R Lacerda
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US.
| |
Collapse
|
29
|
Li T, Qiu J, Jia T, Liang Y, Zhang K, Yan W, Hou Z, Yang S, Liu L, Xiong W, Chen Y, Wang G. G3BP2 regulates oscillatory shear stress-induced endothelial dysfunction. Genes Dis 2021; 9:1701-1715. [PMID: 36157502 PMCID: PMC9485288 DOI: 10.1016/j.gendis.2021.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/06/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
GTPase-activating SH3 domain-binding protein 2 (G3BP2) is a mediator that responds to environmental stresses through stress granule formation and is involved in the progression of chronic diseases. However, no studies have examined the contribution of G3BP2 in the oscillatory shear stress (OSS)-induced endothelial dysfunction. Here we assessed the effects of G3BP2 in endothelial cells (ECs) function and investigated the underlying mechanism. Using shear stress apparatus and partial ligation model, we identified that stress granule-related genes in ECs could be induced by OSS with RNA-seq, and then confirmed that G3BP2 was highly and specifically expressed in athero-susceptible endothelia in the OSS regions. G3bp2–/–Apoe–/– mice had significantly decreased atherosclerotic lesions associated with deficiency of G3BP2 in protecting endothelial barrier function, decreasing monocyte adhesion to ECs and inhibiting the proinflammatory cytokine levels. Furthermore, loss of G3BP2 diminished OSS-induced inflammation in ECs by increasing YAP nucleocytoplasmic shuttling and phosphorylation. These data demonstrate that G3BP2 is a critical OSS regulated gene in regulating ECs function and that G3BP2 inhibition in ECs is a promising atheroprotective therapeutic strategy.
Collapse
Affiliation(s)
- Tianhan Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 45003, PR China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
- Corresponding author.
| | - Tingting Jia
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 45003, PR China
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 45003, PR China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
| | - Wenhua Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
| | - Zhengjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
| | - Shiwei Yang
- Department of Vascular Surgery, First Affiliated Hospital, Army Medical University (Third Military University), Chongqing 400038, PR China
| | - Lushan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Wenhao Xiong
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Yaokai Chen
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing 400030, PR China
- Corresponding author.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State Key Laboratory of Mechanical Transmission, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, PR China
- Corresponding author.
| |
Collapse
|
30
|
Deb N, Ali MS, Mathews A, Chang YW, Lacerda CM. Shear type and magnitude affect aortic valve endothelial cell morphology, orientation, and differentiation. Exp Biol Med (Maywood) 2021; 246:2278-2289. [PMID: 34260291 DOI: 10.1177/15353702211023359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Valvular endothelial cells line the outer layer of heart valves and can withstand shear forces caused by blood flow. In contrast to vascular endothelial cells, there is limited amount of research over valvular endothelial cells. For this reason, the exact physiologic behavior of valvular endothelial cells is unclear. Prior studies have concluded that valvular endothelial cells align perpendicularly to the direction of blood flow, while vascular endothelial cells align parallel to blood flow. Other studies have suggested that different ranges of shear stress uniquely impact the behavior of valvular endothelial cells. The goal of this study was to characterize the response of valvular endothelial cell under different types, magnitudes, and durations of shear stress. In this work, the results demonstrated that with increased shear rate and duration of exposure, valvular endothelial cells no longer possessed the traditional cuboidal morphology. Instead through the change in cell circularity and aspect ratio, valvular endothelial cells aligned in an organized manner. In addition, different forms of shear exposure caused the area and circularity of valvular endothelial cells to decrease while inducing mesenchymal transformation validated through αSMA and TGFβ1 expression. This is the first investigation showing that valvular endothelial cells alignment is not as straightforward as once thought (perpendicular to flow). Different types and magnitudes of shear induce different local behaviors. This is also the first demonstration of valvular endothelial cells undergoing EndMT without chemical inducers on a soft surface in vitro. Findings from this study provide insights to understanding the pathophysiology of valvular endothelial cells which can potentially propel future artificial engineered heart valves.
Collapse
Affiliation(s)
- Nandini Deb
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Mir S Ali
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ashley Mathews
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Ya-Wen Chang
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Carla Mr Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| |
Collapse
|
31
|
The Entry and Egress of Monocytes in Atherosclerosis: A Biochemical and Biomechanical Driven Process. Cardiovasc Ther 2021; 2021:6642927. [PMID: 34345249 PMCID: PMC8282391 DOI: 10.1155/2021/6642927] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
In accordance with “the response to injury” theory, the entry of monocytes into the intima guided by inflammation signals, taking up cholesterol and transforming into foam cells, and egress from plaques determines the progression of atherosclerosis. Multiple cytokines and receptors have been reported to be involved in monocyte recruitment such as CCL2/CCR2, CCL5/CCR5, and CX3CL1/CX3CR1, and the egress of macrophages from the plaque like CCR7/CCL19/CCL21. Interestingly, some neural guidance molecules such as Netrin-1 and Semaphorin 3E have been demonstrated to show an inhibitory effect on monocyte migration. During the processes of monocytes recruitment and migration, factors affecting the biomechanical properties (e.g., the membrane fluidity, the deformability, and stiffness) of the monocytes, like cholesterol, amyloid-β peptide (Aβ), and lipopolysaccharides (LPS), as well as the biomechanical environment that the monocytes are exposed, like the extracellular matrix stiffness, mechanical stretch, blood flow, and hypertension, were discussed in the latter section. Till now, several small interfering RNAs (siRNAs), monoclonal antibodies, and antagonists for CCR2 have been designed and shown promising efficiency on atherosclerosis therapy. Seeking more possible biochemical factors that are chemotactic or can affect the biomechanical properties of monocytes, and uncovering the underlying mechanism, will be helpful in future studies.
Collapse
|
32
|
Left Ventricular Assist Device Support-Induced Alteration of Mechanical Stress on Aortic Valve and Aortic Wall. ASAIO J 2021; 68:516-523. [PMID: 34261872 DOI: 10.1097/mat.0000000000001522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
The aim of this study was to evaluate the fluid dynamics in the aortic valve and proximal aorta during continuous-flow left ventricular assist device (LVAD) support using epiaortic echocardiography and vector flow mapping technology. A total of 12 patients who underwent HeartMate 3 implantation between December 2018 and February 2020 were prospectively examined. The wall shear stress (WSS) on the ascending aorta, aortic root, and aortic valve was evaluated before and after LVAD implantation. The median age of the cohort was 62 years and 17% were women. The peak WSS on the ascending aorta (Pre 1.48 [0.86-1.69] [Pascal {Pa}] vs. Post 0.33 [0.21-0.58] [Pa]; p = 0.002), aortic root (Pre 0.46 [0.31-0.58] (Pa) vs. Post 0.18 [0.12-0.25] (Pa); p = 0.001), and ventricularis of the aortic valve (Pre 1.76 [1.59-2.30] (Pa) vs. Post 0.30 [0.10-0.61] (Pa); p = 0.001) was significantly lower after LVAD implantation. No difference in WSS was observed on the fibrosa of the aortic valve (Pre 0.36 [0.22-0.53] (Pa) vs. Post 0.38 [0.38-0.52] (Pa); p = 0.850) before and after implantation. The WSS on the ascending aorta, aortic root, and ventricularis of the aortic valve leaflets was significantly altered by LVAD implantation, providing preliminary data on the potential contribution of fluid dynamics to LVAD-induced aortic insufficiency and root thrombus.
Collapse
|
33
|
Richfield O, Cortez R, Navar LG. Simulations of Glomerular Shear and Hoop Stresses in Diabetes, Hypertension, and Reduced Renal Mass using a Network Model of a Rat Glomerulus. Physiol Rep 2021; 8:e14577. [PMID: 32951361 PMCID: PMC7507384 DOI: 10.14814/phy2.14577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
A novel anatomically accurate model of rat glomerular filtration is used to quantify shear stresses on the glomerular capillary endothelium and hoop stresses on the glomerular capillary walls. Plasma, erythrocyte volume, and plasma protein mass are distributed at network nodes using pressure differentials calculated taking into account volume loss to filtration, improving on previous models which only took into account blood apparent viscosity in calculating pressures throughout the network. Filtration is found to be heterogeneously distributed throughout the glomerular capillary network and is determined by concentration of plasma proteins and surface area of the filtering capillary segments. Hoop stress is primarily concentrated near the afferent arteriole, whereas shear stress is concentrated near the efferent arteriole. Using parameters from glomerular micropuncture studies, conditions of diabetes mellitus (DM), 5/6‐Nephrectomy (5/6‐Nx), and Angiotensin II‐induced hypertension (HTN) are simulated and compared to their own internal controls to assess the changes in mechanical stresses. Hoop stress is increased in all three conditions, while shear stress is increased in 5/6‐Nx, decreased in HTN, and maintained at control levels in DM by the hypertrophic response of the glomerular capillaries. The results indicate that these alterations in mechanical stresses and the consequent release of cytokines by or injury of the glomerular cells may play a significant role in the progression of glomerulopathy in these disease conditions.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation PhD Program, Tulane University, New Orleans, LA, USA.,Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA, USA
| | - L Gabriel Navar
- Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| |
Collapse
|
34
|
Afshar M, Yazdan-Ashoori S, Engert JC, Thanassoulis G. Drugs for Prevention and Treatment of Aortic Stenosis: How Close Are We? Can J Cardiol 2021; 37:1016-1026. [DOI: 10.1016/j.cjca.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
|
35
|
Huang J, Pu Y, Zhang H, Xie L, He L, Zhang CL, Cheng CK, Huo Y, Wan S, Chen S, Huang Y, Lau CW, Wang L, Xia Y, Huang Y, Luo JY. KLF2 Mediates the Suppressive Effect of Laminar Flow on Vascular Calcification by Inhibiting Endothelial BMP/SMAD1/5 Signaling. Circ Res 2021; 129:e87-e100. [PMID: 34157851 DOI: 10.1161/circresaha.120.318690] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Juan Huang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, China (J.H.)
| | - Yujie Pu
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital (H.Z., S.C.), Nanjing Medical University, China
| | - Liping Xie
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine (L.X.), Nanjing Medical University, China
| | - Lei He
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Cheng-Lin Zhang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Yingsong Huo
- Department of Radiology, Nanjing First Hospital (Y.H.), Nanjing Medical University, China
| | - Song Wan
- Department of Surgery (S.W.), Chinese University of Hong Kong, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital (H.Z., S.C.), Nanjing Medical University, China
| | - Yuhong Huang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Li Wang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Yin Xia
- School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Yu Huang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| | - Jiang-Yun Luo
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China.,School of Biomedical Sciences (J.H., Y.P., L.H., C.-L.Z., C.K.C., Yuhong Huang, C.W.L., L.W., Y.X., Yu Huang, J.-Y.L.), Chinese University of Hong Kong, China
| |
Collapse
|
36
|
Kazik HB, Kandail HS, LaDisa JF, Lincoln J. Molecular and Mechanical Mechanisms of Calcification Pathology Induced by Bicuspid Aortic Valve Abnormalities. Front Cardiovasc Med 2021; 8:677977. [PMID: 34124206 PMCID: PMC8187581 DOI: 10.3389/fcvm.2021.677977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/29/2021] [Indexed: 11/13/2022] Open
Abstract
Bicuspid aortic valve (BAV) is a congenital defect affecting 1-2% of the general population that is distinguished from the normal tricuspid aortic valve (TAV) by the existence of two, rather than three, functional leaflets (or cusps). BAV presents in different morphologic phenotypes based on the configuration of cusp fusion. The most common phenotypes are Type 1 (containing one raphe), where fusion between right coronary and left coronary cusps (BAV R/L) is the most common configuration followed by fusion between right coronary and non-coronary cusps (BAV R/NC). While anatomically different, BAV R/L and BAV R/NC configurations are both associated with abnormal hemodynamic and biomechanical environments. The natural history of BAV has shown that it is not necessarily the primary structural malformation that enforces the need for treatment in young adults, but the secondary onset of premature calcification in ~50% of BAV patients, that can lead to aortic stenosis. While an underlying genetic basis is a major pathogenic contributor of the structural malformation, recent studies have implemented computational models, cardiac imaging studies, and bench-top methods to reveal BAV-associated hemodynamic and biomechanical alterations that likely contribute to secondary complications. Contributions to the field, however, lack support for a direct link between the external valvular environment and calcific aortic valve disease in the setting of BAV R/L and R/NC BAV. Here we review the literature of BAV hemodynamics and biomechanics and discuss its previously proposed contribution to calcification. We also offer means to improve upon previous studies in order to further characterize BAV and its secondary complications.
Collapse
Affiliation(s)
- Hail B. Kazik
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - John F. LaDisa
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Section of Pediatric Cardiology, The Herma Heart Institute, Children's Wisconsin, Milwaukee, WI, United States
| | - Joy Lincoln
- Section of Pediatric Cardiology, The Herma Heart Institute, Children's Wisconsin, Milwaukee, WI, United States
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
37
|
Hakami NY, Dusting GJ, Chan EC, Shah MH, Peshavariya HM. Wound Healing After Alkali Burn Injury of the Cornea Involves Nox4-Type NADPH Oxidase. Invest Ophthalmol Vis Sci 2021; 61:20. [PMID: 33079994 PMCID: PMC7585390 DOI: 10.1167/iovs.61.12.20] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Corneal injury that occurs after burning with alkali initiates wound-healing processes, including inflammation, neovascularization, and fibrosis. Excessive reactions to injury can reduce corneal transparency and thereby compromise vision. The NADPH oxidase (Nox) enzyme complex is known to be involved in cell signaling for wound-healing angiogenesis, but its role in corneal neovascularization has been little studied. Methods The center corneas of wild-type and Nox4 knockout (KO) mice were injured with 3 µL 1 M NaOH, while the contralateral corneas remained untouched. On day 7, mRNA expression levels of NADPH oxidase isoforms, the proangiogenic factors VEGF-A and TGFβ1, and proinflammatory genes ICAM-1 and VCAM-1 were determined. Corneal neovascularization and fibrosis were visualized using PECAM-1 antibody and picrosirius red staining, respectively, on the same day. Results Expressions of both Nox2 and Nox4 gene isoforms as well as the above genes were markedly increased in the injured corneas at 7 days. Injured corneas showed neovascularization and fibrosis as well as an increase in clinical opacity score. All responses stimulated by alkali burn were abrogated in Nox4 KO mice. Conclusions Nox4 could be a new target to treat pathologic corneal wound-healing responses and such targeting might prevent blindness caused by burn injuries.
Collapse
Affiliation(s)
- Nora Y Hakami
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia.,Faculty of Applied Medical Sciences, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Gregory J Dusting
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Elsa C Chan
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Manisha H Shah
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Hitesh M Peshavariya
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Driscoll K, Cruz AD, Butcher JT. Inflammatory and Biomechanical Drivers of Endothelial-Interstitial Interactions in Calcific Aortic Valve Disease. Circ Res 2021; 128:1344-1370. [PMID: 33914601 DOI: 10.1161/circresaha.121.318011] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Calcific aortic valve disease is dramatically increasing in global burden, yet no therapy exists outside of prosthetic replacement. The increasing proportion of younger and more active patients mandates alternative therapies. Studies suggest a window of opportunity for biologically based diagnostics and therapeutics to alleviate or delay calcific aortic valve disease progression. Advancement, however, has been hampered by limited understanding of the complex mechanisms driving calcific aortic valve disease initiation and progression towards clinically relevant interventions.
Collapse
Affiliation(s)
| | - Alexander D Cruz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca NY
| | | |
Collapse
|
39
|
Niazy N, Barth M, Selig JI, Feichtner S, Shakiba B, Candan A, Albert A, Preuß K, Lichtenberg A, Akhyari P. Degeneration of Aortic Valves in a Bioreactor System with Pulsatile Flow. Biomedicines 2021; 9:biomedicines9050462. [PMID: 33922670 PMCID: PMC8145810 DOI: 10.3390/biomedicines9050462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease is the most common valvular heart disease in industrialized countries. Pulsatile pressure, sheer and bending stress promote initiation and progression of aortic valve degeneration. The aim of this work is to establish an ex vivo model to study the therein involved processes. Ovine aortic roots bearing aortic valve leaflets were cultivated in an elaborated bioreactor system with pulsatile flow, physiological temperature, and controlled pressure and pH values. Standard and pro-degenerative treatment were studied regarding the impact on morphology, calcification, and gene expression. In particular, differentiation, matrix remodeling, and degeneration were also compared to a static cultivation model. Bioreactor cultivation led to shrinking and thickening of the valve leaflets compared to native leaflets while gross morphology and the presence of valvular interstitial cells were preserved. Degenerative conditions induced considerable leaflet calcification. In comparison to static cultivation, collagen gene expression was stable under bioreactor cultivation, whereas expression of hypoxia-related markers was increased. Osteopontin gene expression was differentially altered compared to protein expression, indicating an enhanced protein turnover. The present ex vivo model is an adequate and effective system to analyze aortic valve degeneration under controlled physiological conditions without the need of additional growth factors.
Collapse
Affiliation(s)
- Naima Niazy
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Sabine Feichtner
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Babak Shakiba
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Asya Candan
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Alexander Albert
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
- Department of Cardiovascular Surgery, Klinikum Dortmund gGmbH, Beurhausstraße 40, 44137 Dortmund, Germany
| | - Karlheinz Preuß
- Faculty of Biotechnology, Bioprocessing, Modulation and Simulation, University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163 Mannheim, Germany;
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
- Correspondence:
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| |
Collapse
|
40
|
Hayashi H, Itatani K, Akiyama K, Zhao Y, Kurlansky P, DeRoo S, Sanchez J, Ferrari G, Yuzefpolskaya M, Colombo PC, Takeda K, Wu IY, Kainuma A, Takayama H. Influence of aneurysmal aortic root geometry on mechanical stress to the aortic valve leaflet. Eur Heart J Cardiovasc Imaging 2021; 22:986-994. [PMID: 33611382 DOI: 10.1093/ehjci/jeab006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/08/2021] [Indexed: 01/16/2023] Open
Abstract
AIMS While mechanical stress caused by blood flow, e.g. wall shear stress (WSS), and related parameters, e.g. oscillatory shear index (OSI), are increasingly being recognized as key moderators of various cardiovascular diseases, studies on valves have been limited because of a lack of appropriate imaging modalities. We investigated the influence of aortic root geometry on WSS and OSI on the aortic valve (AV) leaflet. METHODS AND RESULTS We applied our novel approach of intraoperative epi-aortic echocardiogram to measure the haemodynamic parameters of WSS and OSI on the AV leaflet. Thirty-six patients were included, which included those who underwent valve-sparing aortic root replacement (VSARR) with no significant aortic regurgitation (n = 17) and coronary artery bypass graft (CABG) with normal AV (n = 19). At baseline, those who underwent VSARR had a higher systolic WSS (0.52 ± 0.12 vs. 0.32 ± 0.08 Pa, respectively, P < 0.001) and a higher OSI (0.37 ± 0.06 vs. 0.29 ± 0.04, respectively, P < 0.001) on the aortic side of the AV leaflet than those who underwent CABG. Multivariate regression analysis revealed that the size of the sinus of Valsalva had a significant association with WSS and OSI. Following VSARR, WSS and OSI values decreased significantly compared with the baseline values (WSS: 0.29 ± 0.12 Pa, P < 0.001; OSI: 0.26 ± 0.09, P < 0.001), and became comparable to the values in those who underwent CABG (WSS, P = 0.42; OSI, P = 0.15). CONCLUSIONS Mechanical stress on the AV gets altered in correlation with the size of the aortic root. An aneurysmal aortic root may expose the leaflet to abnormal fluid dynamics. The VSARR procedure appeared to reduce these abnormalities.
Collapse
Affiliation(s)
- Hideyuki Hayashi
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Keiichi Itatani
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Koichi Akiyama
- Department of Anesthesiology, Yodogawa Christian Hospital, 1 Chome-7-50, Kunijima, Higashiyodogawa Ward, Osaka, 533-0024, Japan
| | - Yanling Zhao
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Paul Kurlansky
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Scott DeRoo
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Joseph Sanchez
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Giovanni Ferrari
- Department of Surgery and Biomedical Engineering, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Melana Yuzefpolskaya
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Koji Takeda
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Isaac Y Wu
- Department of Anesthesiology, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Atsushi Kainuma
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| | - Hiroo Takayama
- Division of Cardiothoracic Surgery, Department of Surgery, New York-Presbyterian Hospital, Columbia University Medical Center, 707 Fort Washington Avenue, New York, NY 10032, USA
| |
Collapse
|
41
|
Shar JA, Keswani SG, Grande-Allen KJ, Sucosky P. Computational Assessment of Valvular Dysfunction in Discrete Subaortic Stenosis: A Parametric Study. Cardiovasc Eng Technol 2021; 12:559-575. [PMID: 33432514 DOI: 10.1007/s13239-020-00513-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/22/2020] [Indexed: 01/16/2023]
Abstract
PURPOSE Discrete subaortic stenosis (DSS) is a left-ventricular outflow tract (LVOT) obstruction caused by a membranous lesion. DSS is associated with steep aortoseptal angles (AoSAs) and is a risk factor for aortic regurgitation (AR). However, the etiology of AR secondary to DSS remains unknown. This study aimed at quantifying computationally the impact of AoSA steepening and DSS on aortic valve (AV) hemodynamics and AR. METHODS An LV geometry reconstructed from cine-MRI data was connected to an AV geometry to generate a unified 2D LV-AV model. Six geometrical variants were considered: unobstructed (CTRL) and DSS-obstructed LVOT (DSS), each reflecting three AoSA variations (110°, 120°, 130°). Fluid-structure interaction simulations were run to compute LVOT flow, AV leaflet dynamics, and regurgitant fraction (RF). RESULTS AoSA steepening and DSS generated vortex dynamics alterations and stenotic flow conditions. While the CTRL-110° model generated the highest degree of leaflet opening asymmetry, DSS preferentially altered superior leaflet kinematics, and caused leaflet-dependent alterations in systolic fluttering. LVOT steepening and DSS subjected the leaflets to increasing WSS overloads (up to 94% increase in temporal shear magnitude), while DSS also increased WSS bidirectionality on the inferior leaflet belly (+ 0.30-point in oscillatory shear index). Although AoSA steepening and DSS increased diastolic transvalvular backflow, regurgitant fractions (RF < 7%) remained below the threshold defining clinical mild AR. CONCLUSIONS The mechanical interactions between AV leaflets and LVOT steepening/DSS hemodynamic derangements do not cause AR. However, the leaflet WSS abnormalities predicted in those anatomies provide new support to a mechanobiological etiology of AR secondary to DSS.
Collapse
Affiliation(s)
- Jason A Shar
- Department of Mechanical and Materials Engineering, Wright State University, Dayton, USA
| | - Sundeep G Keswani
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Baylor College of Medicine, Houston, USA
| | | | - Philippe Sucosky
- Department of Mechanical Engineering, Kennesaw State University, 840 Polytechnic Lane, Marietta, GA, 30060, USA.
| |
Collapse
|
42
|
Flemister DC, Hatoum H, Guhan V, Zebhi B, Lincoln J, Crestanello J, Dasi LP. Effect of Left and Right Coronary Flow Waveforms on Aortic Sinus Hemodynamics and Leaflet Shear Stress: Correlation with Calcification Locations. Ann Biomed Eng 2020; 48:2796-2808. [PMID: 33145675 PMCID: PMC11022940 DOI: 10.1007/s10439-020-02677-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022]
Abstract
Coronary flow induces hemodynamic alterations in the aortic sinus region. The objectives of this study are to: (1) investigate the differences among sinus hemodynamics and leaflet wall shear stresses engendered by the left versus right versus non-coronary flow and (2) correlate respective wall shear stresses with leaflet calcification in patients. A left heart simulator flow loop with a tunable coronary circuit provided physiological coronary flow waveforms corresponding to the left coronary cusp case (LCC), right coronary cusp case (RCC), and non-coronary cusp case (NCC). High spatio-temporal resolution particle image velocimetry was conducted to quantify leaflet wall shear stress and sinus vorticity fields and to measure aortic leaflet tip kinematics. Thirty-one patients with severe calcific aortic valve disease were segmented from CT data for the calcific volumes in their respective left, right, and non-coronary cusps. Leaflet tip position during systole shows the RCC has a wider leaflet opening compared to LCC and NCC. Velocity and vorticity fields combined with leaflet position data show that sinus vorticity is diminished (peak ~ 43 s-1) in the LCC while RCC and NCC maintain high vorticity (~ 1200 and ~ 950 s-1 respectively). WSS magnitudes greater than 0.3 Pa show 20 and 81% greater occurrences in the LCC and RCC respectively compared to NCC. Significant differences [X2 (2, n = 31) = 7.31, p = 0.0258] between the calcification levels in each cusp of the patient population. Coronary flow differences between LCC, RCC, and NCC show significant impact on leaflet kinematics and sinus flow hemodynamics. Clinical data correlations of the coronary flow cases indicate the left coronary cusp has a higher likelihood of calcification compared to the right.
Collapse
Affiliation(s)
- Dorma C Flemister
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Hoda Hatoum
- Department of Biomedical Engineering, Georgia Institute of Technology, 387 Technology Circle, Atlanta, GA, 30313, USA
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931, USA
| | - Varshini Guhan
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Joy Lincoln
- Department of Pediatric Cardiology, Children's Hospital of Wisconsin, Milwaukee, WI, USA
| | | | - Lakshmi P Dasi
- Department of Biomedical Engineering, Georgia Institute of Technology, 387 Technology Circle, Atlanta, GA, 30313, USA.
| |
Collapse
|
43
|
Hsu CPD, Hutcheson JD, Ramaswamy S. Oscillatory fluid-induced mechanobiology in heart valves with parallels to the vasculature. VASCULAR BIOLOGY 2020; 2:R59-R71. [PMID: 32923975 PMCID: PMC7439923 DOI: 10.1530/vb-19-0031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/17/2020] [Indexed: 12/31/2022]
Abstract
Forces generated by blood flow are known to contribute to cardiovascular development and remodeling. These hemodynamic forces induce molecular signals that are communicated from the endothelium to various cell types. The cardiovascular system consists of the heart and the vasculature, and together they deliver nutrients throughout the body. While heart valves and blood vessels experience different environmental forces and differ in morphology as well as cell types, they both can undergo pathological remodeling and become susceptible to calcification. In addition, while the plaque morphology is similar in valvular and vascular diseases, therapeutic targets available for the latter condition are not effective in the management of heart valve calcification. Therefore, research in valvular and vascular pathologies and treatments have largely remained independent. Nonetheless, understanding the similarities and differences in development, calcific/fibrous pathologies and healthy remodeling events between the valvular and vascular systems can help us better identify future treatments for both types of tissues, particularly for heart valve pathologies which have been understudied in comparison to arterial diseases.
Collapse
Affiliation(s)
- Chia-Pei Denise Hsu
- Engineering Center, Department of Biomedical Engineering, Florida International University, Miami, Florida, USA
| | - Joshua D Hutcheson
- Engineering Center, Department of Biomedical Engineering, Florida International University, Miami, Florida, USA
| | - Sharan Ramaswamy
- Engineering Center, Department of Biomedical Engineering, Florida International University, Miami, Florida, USA
| |
Collapse
|
44
|
Side-dependent effect in the response of valve endothelial cells to bidirectional shear stress. Int J Cardiol 2020; 323:220-228. [PMID: 32858136 DOI: 10.1016/j.ijcard.2020.08.074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
Abstract
Endothelial cells covering the aortic and ventricular sides of the aortic valve leaflets are exposed to different stresses, in particular wall shear stress (WSS). Biomechanical stimuli actively regulate valve tissue structure and induce remodeling events leading to valve dysfunction. Endothelial to mesenchymal transformation (EndMT), for example, has been associated with aortic valve disease. The biomechanical response of cells at different sides of the leaflets has not been clearly characterized. To analyze the mechanical response of valve endothelial cells (VECs) we developed a unique fluid activation device that applies physiologically relevant pulsatile WSS. We characterized the morphology and function of adult porcine aortic VECs derived from the opposite sides of aortic valve leaflets following exposure to different pulsatile WSS. We found that elongation and orientation of cells in response to pulsatile WSS depends on their side of origin. Quantification of gene expression confirms phenotypic differences between aortic and ventricular VECs. Aortic VECs exposed to pulsatile WSS similar to that in vivo at the tip of aortic side of the valve leaflet upregulated pro-EndMT (ACTA2, Snail, TGFβ1) and inflammation (ICAM-1, VCAM-1) genes, whereas expression of endothelial markers like PECAM-1 was decreased. Conversely, ventricular-VECs showed strong increase of PECAM-1 expression and no activation of pro-EndMT marker. Finally, we found that stress-induced genes are upregulated in both cell types, at higher levels in ventricular compared to aortic VECs. Application of physiological shear stress levels using a fluid activation device therefore reveals functional differences in VECs derived from opposite sides of the aortic valve leaflets.
Collapse
|
45
|
Emendi M, Sturla F, Ghosh RP, Bianchi M, Piatti F, Pluchinotta FR, Giese D, Lombardi M, Redaelli A, Bluestein D. Patient-Specific Bicuspid Aortic Valve Biomechanics: A Magnetic Resonance Imaging Integrated Fluid-Structure Interaction Approach. Ann Biomed Eng 2020; 49:627-641. [PMID: 32804291 DOI: 10.1007/s10439-020-02571-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022]
Abstract
Congenital bicuspid aortic valve (BAV) consists of two fused cusps and represents a major risk factor for calcific valvular stenosis. Herein, a fully coupled fluid-structure interaction (FSI) BAV model was developed from patient-specific magnetic resonance imaging (MRI) and compared against in vivo 4-dimensional flow MRI (4D Flow). FSI simulation compared well with 4D Flow, confirming direction and magnitude of the flow jet impinging onto the aortic wall as well as location and extension of secondary flows and vortices developing at systole: the systolic flow jet originating from an elliptical 1.6 cm2 orifice reached a peak velocity of 252.2 cm/s, 0.6% lower than 4D Flow, progressively impinging on the ascending aorta convexity. The FSI model predicted a peak flow rate of 22.4 L/min, 6.7% higher than 4D Flow, and provided BAV leaflets mechanical and flow-induced shear stresses, not directly attainable from MRI. At systole, the ventricular side of the non-fused leaflet revealed the highest wall shear stress (WSS) average magnitude, up to 14.6 Pa along the free margin, with WSS progressively decreasing towards the belly. During diastole, the aortic side of the fused leaflet exhibited the highest diastolic maximum principal stress, up to 322 kPa within the attachment region. Systematic comparison with ground-truth non-invasive MRI can improve the computational model ability to reproduce native BAV hemodynamics and biomechanical response more realistically, and shed light on their role in BAV patients' risk for developing complications; this approach may further contribute to the validation of advanced FSI simulations designed to assess BAV biomechanics.
Collapse
Affiliation(s)
- Monica Emendi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Francesco Sturla
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Ram P Ghosh
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Matteo Bianchi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Filippo Piatti
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Francesca R Pluchinotta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.,Multimodality Cardiac Imaging, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.,Department of Pediatric and Adult Congenital Heart Disease, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | | | - Massimo Lombardi
- Multimodality Cardiac Imaging, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
46
|
Wang J, Zhang S. Fluid shear stress modulates endothelial inflammation by targeting LIMS2. Exp Biol Med (Maywood) 2020; 245:1656-1663. [PMID: 32752897 DOI: 10.1177/1535370220943837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mechanosensitive genes regulate multiple cardiovascular pathophysiological processes and disorders; however, the role of flow-sensitive genes in atherosclerosis is still unknown. In this study, we identify LIM Zinc Finger Domain Containing 2 (LIMS2) that acts as a mechanosensitive gene downregulated by disturbed flow (d-flow) both in human endothelial cells (ECs) in vitro and in mice in vivo. Mechanistically, d-flow suppresses LIMS2 expression, which leads to endothelial inflammation by upregulating typical inflammatory factors, VCAM-1, and ICAM-1 in human ECs. The findings indicate that LIMS2, the new flow-sensitive gene, may help us to find a new insight to explain how d-flow caused endothelial inflammation and provide a new therapeutic approach for atherosclerosis in the future.
Collapse
Affiliation(s)
- Junyao Wang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shiyanjin Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
47
|
Teng P, Xu X, Ni C, Yan H, Sun Q, Zhang E, Ni Y. Identification of key genes in calcific aortic valve disease by integrated bioinformatics analysis. Medicine (Baltimore) 2020; 99:e21286. [PMID: 32702920 PMCID: PMC7373610 DOI: 10.1097/md.0000000000021286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is highly prevalent in our aging world and has no effective pharmaceutical treatment. Intense efforts have been made but the underlying molecular mechanisms of CAVD are still unclear.This study was designed to identify the critical genes and pathways in CAVD by bioinformatics analysis. Microarray datasets of GSE12644, GSE51472, and GSE83453 were obtained from Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified and functional and pathway enrichment analysis was performed. Subsequently, the protein-protein interaction network (PPI) was constructed with Search Tool for the Retrieval of Interacting Genes and was visualized with Cytoscape to identify the most significant module. Hub genes were identified by Cytoscape plugin cytoHubba.A total of 179 DEGs, including 101 upregulated genes and 78 downregulated genes, were identified. The enriched functions and pathways of the DEGs include inflammatory and immune response, chemotaxis, extracellular matrix (ECM) organization, complement and coagulation cascades, ECM receptor interaction, and focal adhesion. The most significant module in the PPI network was analyzed and genes among it were mainly enriched in chemotaxis, locomotory behavior, immune response, chemokine signaling pathway, and extracellular space. In addition, DEGs, with degrees ≥ 10 and the top 10 highest Maximal Chique Centrality (MCC) score, were identified as hub genes. CCR1, MMP9, VCAM1, and ITGAX, which were of the highest degree or MCC score, were manually reviewed.The DEGs and hub genes identified in the present study help us understand the molecular mechanisms underlying the pathogenesis of CAVD and might serve as candidate therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Peng Teng
- Department of Cardiothoracic Surgery
| | | | | | - Haimeng Yan
- Department of Bone Marrow Transplantation Center
| | - Qianhui Sun
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Enfan Zhang
- Department of Bone Marrow Transplantation Center
| | - Yiming Ni
- Department of Cardiothoracic Surgery
| |
Collapse
|
48
|
Pathogenic Pathways and Therapeutic Approaches Targeting Inflammation in Diabetic Nephropathy. Int J Mol Sci 2020; 21:ijms21113798. [PMID: 32471207 PMCID: PMC7312633 DOI: 10.3390/ijms21113798] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is associated with an increased morbidity and mortality, resulting in elevated cost for public health systems. DN is the main cause of chronic kidney disease (CKD) and its incidence increases the number of patients that develop the end-stage renal disease (ESRD). There are growing epidemiological and preclinical evidence about the close relationship between inflammatory response and the occurrence and progression of DN. Several anti-inflammatory strategies targeting specific inflammatory mediators (cell adhesion molecules, chemokines and cytokines) and intracellular signaling pathways have shown beneficial effects in experimental models of DN, decreasing proteinuria and renal lesions. A number of inflammatory molecules have been shown useful to identify diabetic patients at high risk of developing renal complications. In this review, we focus on the key role of inflammation in the genesis and progression of DN, with a special interest in effector molecules and activated intracellular pathways leading to renal damage, as well as a comprehensive update of new therapeutic strategies targeting inflammation to prevent and/or retard renal injury.
Collapse
|
49
|
Alushi B, Curini L, Christopher MR, Grubitzch H, Landmesser U, Amedei A, Lauten A. Calcific Aortic Valve Disease-Natural History and Future Therapeutic Strategies. Front Pharmacol 2020; 11:685. [PMID: 32477143 PMCID: PMC7237871 DOI: 10.3389/fphar.2020.00685] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most frequent heart valve disorder. It is characterized by an active remodeling process accompanied with valve mineralization, that results in a progressive aortic valve narrowing, significant restriction of the valvular area, and impairment of blood flow.The pathophysiology of CAVD is a multifaceted process, involving genetic factors, chronic inflammation, lipid deposition, and valve mineralization. Mineralization is strictly related to the inflammatory process in which both, innate, and adaptive immunity are involved. The underlying pathophysiological pathways that go from inflammation to calcification and, finally lead to severe stenosis, remain, however, incompletely understood. Histopathological studies are limited to patients with severe CAVD and no samples are available for longitudinal studies of disease progression. Therefore, alternative routes should be explored to investigate the pathogenesis and progression of CAVD.Recently, increasing evidence suggests that epigenetic markers such as non-coding RNAs are implicated in the landscape of phenotypical changes occurring in CAVD. Furthermore, the microbiome, an essential player in several diseases, including the cardiovascular ones, has recently been linked to the inflammation process occurring in CAVD. In the present review, we analyze and discuss the CAVD pathophysiology and future therapeutic strategies, focusing on the real and putative role of inflammation, calcification, and microbiome.
Collapse
Affiliation(s)
- Brunilda Alushi
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| | - Lavinia Curini
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Mary Roxana Christopher
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Herko Grubitzch
- Berlin Institute of Health, Berlin, Germany.,Department of Cardiology, German Heart Centre Berlin (DHZB), Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,Sod of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Alexander Lauten
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| |
Collapse
|
50
|
Quaglino D, Boraldi F, Lofaro FD. The biology of vascular calcification. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 354:261-353. [PMID: 32475476 DOI: 10.1016/bs.ircmb.2020.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vascular calcification (VC), characterized by different mineral deposits (i.e., carbonate apatite, whitlockite and hydroxyapatite) accumulating in blood vessels and valves, represents a relevant pathological process for the aging population and a life-threatening complication in acquired and in genetic diseases. Similarly to bone remodeling, VC is an actively regulated process in which many cells and molecules play a pivotal role. This review aims at: (i) describing the role of resident and circulating cells, of the extracellular environment and of positive and negative factors in driving the mineralization process; (ii) detailing the types of VC (i.e., intimal, medial and cardiac valve calcification); (iii) analyzing rare genetic diseases underlining the importance of altered pyrophosphate-dependent regulatory mechanisms; (iv) providing therapeutic options and perspectives.
Collapse
Affiliation(s)
- Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | |
Collapse
|