1
|
Plunkett MJ, Paton JFR, Fisher JP. Autonomic control of the pulmonary circulation: Implications for pulmonary hypertension. Exp Physiol 2025; 110:42-57. [PMID: 39453284 DOI: 10.1113/ep092249] [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: 08/15/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024]
Abstract
The autonomic regulation of the pulmonary vasculature has been under-appreciated despite the presence of sympathetic and parasympathetic neural innervation and adrenergic and cholinergic receptors on pulmonary vessels. Recent clinical trials targeting this innervation have demonstrated promising effects in pulmonary hypertension, and in this context of reignited interest, we review autonomic pulmonary vascular regulation, its integration with other pulmonary vascular regulatory mechanisms, systemic homeostatic reflexes and their clinical relevance in pulmonary hypertension. The sympathetic and parasympathetic nervous systems can affect pulmonary vascular tone and pulmonary vascular stiffness. Local afferents in the pulmonary vasculature are activated by elevations in pressure and distension and lead to distinct pulmonary baroreflex responses, including pulmonary vasoconstriction, increased sympathetic outflow, systemic vasoconstriction and increased respiratory drive. Autonomic pulmonary vascular control interacts with, and potentially makes a functional contribution to, systemic homeostatic reflexes, such as the arterial baroreflex. New experimental therapeutic applications, including pulmonary artery denervation, pharmacological cholinergic potentiation, vagal nerve stimulation and carotid baroreflex stimulation, have shown some promise in the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Michael J Plunkett
- Department of Physiology, Faculty of Medical and Health Sciences, Manaaki Manawa - The Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical and Health Sciences, Manaaki Manawa - The Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - James P Fisher
- Department of Physiology, Faculty of Medical and Health Sciences, Manaaki Manawa - The Centre for Heart Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Wong J, Zhao G, Adams-Tzivelekidis S, Wen H, Chandrasekaran P, Michki SN, Gentile ME, Singh M, Kass-Gergi S, Mendoza M, Holcomb NP, Li X, Tang AT, Negretti NM, Sucre JMS, Frank DB, Vaughan AE. Dynamic behavior and lineage plasticity of the pulmonary venous endothelium. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1584-1600. [PMID: 39653825 DOI: 10.1038/s44161-024-00573-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Repair of the pulmonary vascular bed and the origin of new vasculature remain underexplored despite the critical necessity to meet oxygen demands after injury. Given their critical role in angiogenesis in other settings, we investigated the role of venous endothelial cells in endothelial regeneration after adult lung injury. Here we identified Slc6a2 as a marker of pulmonary venous endothelial cells and generated a venous-specific, inducible Cre mouse line. We observed that venous endothelial cells proliferate into the adjacent capillary bed upon influenza injury and hyperoxia injury. Imaging analysis demonstrated that venous endothelial cells proliferate and differentiate into general capillary and aerocyte capillary endothelial cells after infection, thus contributing to repair of the capillary plexus vital for gas exchange. Our studies thus establish that venous endothelial cells exhibit demonstrable progenitor capacity upon respiratory viral injury and sterile injury, contributing to repair of the alveolar capillary bed responsible for pulmonary function.
Collapse
Affiliation(s)
- Joanna Wong
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie Adams-Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongbo Wen
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Prashant Chandrasekaran
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sylvia N Michki
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maria E Gentile
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Madeline Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Kass-Gergi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meryl Mendoza
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas P Holcomb
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinyuan Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas M Negretti
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - David B Frank
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Ji X, Wang L, Zhong Y, Xu Q, Yan J, Pan D, Xu Y, Chen C, Wang J, Wang G, Yang M, Li T, Tang L, Wang X. Impact of mesenchymal stem cell size and adhesion modulation on in vivo distribution: insights from quantitative PET imaging. Stem Cell Res Ther 2024; 15:456. [PMID: 39609885 PMCID: PMC11606219 DOI: 10.1186/s13287-024-04078-4] [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: 07/06/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Successful engraftment and localization of mesenchymal stem cells (MSCs) within target tissues are critical factors influencing their therapeutic efficacy for tissue repair and regeneration. However, the relative contributions of biophysical factors like cell size and adhesion capacity in regulating MSC distribution in vivo remain incompletely understood. METHODS Cell adhesion peptides and hanging drop method were used to modify the adhesive capacity and size of MSCs. To quantitatively track the real-time biodistribution of transplanted MSCs with defined size and adhesion profiles in living mice and rats, the non-invasive positron emission tomography (PET) imaging was applied. RESULTS Surface modification with integrin binding peptides like RGD, GFOGER, and HAVDI reduced MSC adhesion capacity in vitro by up to 43.5% without altering cell size, but did not significantly decrease lung entrapment in vivo. In contrast, culturing MSCs as 3D spheroids for 48 h reduced their cell diameter by 34.6% and markedly enhanced their ability to pass through the lungs and migrate to other organs like the liver after intravenous administration. This size-dependent effect on MSC distribution was more pronounced in rats compared to mice, likely due to differences in pulmonary microvessel diameters between species. CONCLUSION Our findings reveal that cell size is a predominant biophysical regulator of MSC localization in vivo compared to adhesion capacity, providing crucial insights to guide optimization of MSC delivery strategies for enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Xin Ji
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China
| | - Lizhen Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Yudan Zhong
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Qian Xu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Junjie Yan
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Yuping Xu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Chongyang Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd., Nanjing, 211100, P.R. China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Min Yang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Tiannv Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China
| | - Lijun Tang
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China.
| | - Xinyu Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China.
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China.
| |
Collapse
|
4
|
Mukasheva F, Adilova L, Dyussenbinov A, Yernaimanova B, Abilev M, Akilbekova D. Optimizing scaffold pore size for tissue engineering: insights across various tissue types. Front Bioeng Biotechnol 2024; 12:1444986. [PMID: 39600888 PMCID: PMC11588461 DOI: 10.3389/fbioe.2024.1444986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Scaffold porosity is a critical factor in replicating the complex in vivo microenvironment, directly influencing cellular interactions, migration, nutrient transfer, vascularization, and the formation of functional tissues. For optimal tissue formation, scaffold design must account for various parameters, including material composition, morphology, mechanical properties, and cellular compatibility. This review highlights the importance of interconnected porosity and pore size, emphasizing their impact on cellular behavior and tissue formation across several tissue engineering domains, such as skin, bone, cardiovascular, and lung tissues. Specific pore size ranges enhance scaffold functionality for different tissues: small pores (∼1-2 µm) aid epidermal cell attachment in skin regeneration, moderate pores (∼2-12 µm) support dermal migration, and larger pores (∼40-100 µm) facilitate vascular structures. For bone tissue engineering, multi-layered scaffolds with smaller pores (50-100 µm) foster cell attachment, while larger pores (200-400 µm) enhance nutrient diffusion and angiogenesis. Cardiovascular and lung tissues benefit from moderate pore sizes (∼25-60 µm) to balance cell integration and nutrient diffusion. By addressing critical design challenges and optimizing pore size distributions, this review provides insights into scaffold innovations, ultimately advancing tissue regeneration strategies.
Collapse
Affiliation(s)
- Fariza Mukasheva
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Laura Adilova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Aibek Dyussenbinov
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Bota Yernaimanova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| | - Madi Abilev
- Department of Analytical, Colloid Chemistry and Technology of Rare Elements, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Dana Akilbekova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
5
|
Barry M, Barry F, Gun M, Padurean P, Havet E, Gara Ali B, Caus T. Impact of aortic and pulmonary artery wall histology on radicular dilatation during the Ross procedure. J Cardiothorac Surg 2024; 19:618. [PMID: 39487528 PMCID: PMC11531152 DOI: 10.1186/s13019-024-03125-8] [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: 08/09/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024] Open
Abstract
OBJECTIVE In our study, we aim to explore the structural differences between the aortic root and the pulmonary artery to better understand the process of pulmonary autograft dilatation during the Ross procedure. MATERIALS AND METHODS We studied twenty human fetuses (aged 14-36 weeks of gestation) and four adults (one female and three males, aged 30-45 years, mean age = 37 ± 16 years). Samples of aortic root and pulmonary artery were obtained through dissection. Histological examinations, including hematoxylin-eosin, Masson's trichrome, and orcein staining, as well as immunohistochemical technique with caldesmon staining, were performed. Microscopic counting was conducted to assess the number of elastic laminae and smooth muscle cells in each arterial wall. Statistical analyses were performed using R software. Means and standard deviations were used to present central tendencies and data dispersion for elastic laminae and smooth muscle. RESULTS Significant histological differences were observed between the aortic root and pulmonary artery in both adults and fetuses. In fetuses, no difference was found between the two vessels in terms of elastic laminae (p = 0.26) and smooth muscle cells (p = 0.69). However, in adults, significant differences were found for elastic laminae (p < 0.001) and smooth muscle cells (p < 0.001) between the aorta and pulmonary artery. CONCLUSIONS The microscopic vascular structure impacts the mechanical properties of the pulmonary autograft wall and explains its observed dilatation remote from the Ross procedure due to wall stresses related to systemic pressure.
Collapse
Affiliation(s)
- Misbaou Barry
- Department of Cardiac Surgery, Heart-Chest-Vascular Surgery Center, Amiens Picardie University Hospital Center, 1 Rue du Professeur Christian Cabrol, 80054, Amiens, Cedex1, France.
- Anatomy Laboratory, Faculty of Medicine University of Amiens, Amiens, France.
| | - Fatoumata Barry
- Department of Pathological Anatomy and Cytology, Ambroise-Paré Hospital, AP-HP, 9 Avenue Charles-de-Gaulle, 92100, Boulogne-Billancourt, France
| | - Mesut Gun
- Department of Cardiology, Amiens Picardie University Hospital Center, 1 Rue du Professeur Christian Cabrol, 80054, Amiens, Cedex1, France
| | - Paul Padurean
- Department of Cardiac Surgery, Amiens Picardie University Hospital Center, 1 Rue du Professeur Christian Cabrol, 80054, Amiens, Cedex1, France
| | - Eric Havet
- Anatomy Laboratory, Faculty of Medicine University of Amiens, Amiens, France
| | - Bessem Gara Ali
- Department of Cardiac Surgery, Amiens Picardie University Hospital Center, 1 Rue du Professeur Christian Cabrol, 80054, Amiens, Cedex1, France
| | - Thierry Caus
- Department of Cardiac Surgery, Amiens Picardie University Hospital Center, 1 Rue du Professeur Christian Cabrol, 80054, Amiens, Cedex1, France
| |
Collapse
|
6
|
Corboz MR, Nguyen TL, Stautberg A, Cipolla D, Perkins WR, Chapman RW. Current Overview of the Biology and Pharmacology in Sugen/Hypoxia-Induced Pulmonary Hypertension in Rats. J Aerosol Med Pulm Drug Deliv 2024; 37:241-283. [PMID: 39388691 PMCID: PMC11502635 DOI: 10.1089/jamp.2024.0016] [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: 04/03/2024] [Accepted: 07/03/2024] [Indexed: 10/12/2024] Open
Abstract
The Sugen 5416/hypoxia (Su/Hx) rat model of pulmonary arterial hypertension (PAH) demonstrates most of the distinguishing features of PAH in humans, including increased wall thickness and obstruction of the small pulmonary arteries along with plexiform lesion formation. Recently, significant advancement has been made describing the epidemiology, genomics, biochemistry, physiology, and pharmacology in Su/Hx challenge in rats. For example, there are differences in the overall reactivity to Su/Hx challenge in different rat strains and only female rats respond to estrogen treatments. These conditions are also encountered in human subjects with PAH. Also, there is a good translation in both the biochemical and metabolic pathways in the pulmonary vasculature and right heart between Su/Hx rats and humans, particularly during the transition from the adaptive to the nonadaptive phase of right heart failure. Noninvasive techniques such as echocardiography and magnetic resonance imaging have recently been used to evaluate the progression of the pulmonary vascular and cardiac hemodynamics, which are important parameters to monitor the efficacy of drug treatment over time. From a pharmacological perspective, most of the compounds approved clinically for the treatment of PAH are efficacious in Su/Hx rats. Several compounds that show efficacy in Su/Hx rats have advanced into phase II/phase III studies in humans with positive results. Results from these drug trials, if successful, will provide additional treatment options for patients with PAH and will also further validate the excellent translation that currently exists between Su/Hx rats and the human PAH condition.
Collapse
|
7
|
Bartolo MA, Taylor-LaPole AM, Gandhi D, Johnson A, Li Y, Slack E, Stevens I, Turner ZG, Weigand JD, Puelz C, Husmeier D, Olufsen MS. Computational framework for the generation of one-dimensional vascular models accounting for uncertainty in networks extracted from medical images. J Physiol 2024; 602:3929-3954. [PMID: 39075725 DOI: 10.1113/jp286193] [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: 12/22/2023] [Accepted: 05/28/2024] [Indexed: 07/31/2024] Open
Abstract
One-dimensional (1D) cardiovascular models offer a non-invasive method to answer medical questions, including predictions of wave-reflection, shear stress, functional flow reserve, vascular resistance and compliance. This model type can predict patient-specific outcomes by solving 1D fluid dynamics equations in geometric networks extracted from medical images. However, the inherent uncertainty in in vivo imaging introduces variability in network size and vessel dimensions, affecting haemodynamic predictions. Understanding the influence of variation in image-derived properties is essential to assess the fidelity of model predictions. Numerous programs exist to render three-dimensional surfaces and construct vessel centrelines. Still, there is no exact way to generate vascular trees from the centrelines while accounting for uncertainty in data. This study introduces an innovative framework employing statistical change point analysis to generate labelled trees that encode vessel dimensions and their associated uncertainty from medical images. To test this framework, we explore the impact of uncertainty in 1D haemodynamic predictions in a systemic and pulmonary arterial network. Simulations explore haemodynamic variations resulting from changes in vessel dimensions and segmentation; the latter is achieved by analysing multiple segmentations of the same images. Results demonstrate the importance of accurately defining vessel radii and lengths when generating high-fidelity patient-specific haemodynamics models. KEY POINTS: This study introduces novel algorithms for generating labelled directed trees from medical images, focusing on accurate junction node placement and radius extraction using change points to provide haemodynamic predictions with uncertainty within expected measurement error. Geometric features, such as vessel dimension (length and radius) and network size, significantly impact pressure and flow predictions in both pulmonary and aortic arterial networks. Standardizing networks to a consistent number of vessels is crucial for meaningful comparisons and decreases haemodynamic uncertainty. Change points are valuable to understanding structural transitions in vascular data, providing an automated and efficient way to detect shifts in vessel characteristics and ensure reliable extraction of representative vessel radii.
Collapse
Affiliation(s)
- Michelle A Bartolo
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
| | | | - Darsh Gandhi
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
- Department of Mathematics, University of Texas at Arlington, Arlington, TX, USA
| | - Alexandria Johnson
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
- Department of Mathematics and Statistics, University of South Florida, Tampa, FL, USA
| | - Yaqi Li
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
- North Carolina School of Science and Mathematics, Durham, NC, USA
| | - Emma Slack
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
- Department of Mathematics, Colorado State University, Fort Collins, CO, USA
| | - Isaiah Stevens
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
| | - Zachary G Turner
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, USA
| | - Justin D Weigand
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Charles Puelz
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Dirk Husmeier
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Mette S Olufsen
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
8
|
Rivera SI, Bernal CP, Martínez-Peláez R, Robledo-Nolasco R, De León-Larios G, Félix VG, Ostos R, Maestre GE, Melgarejo JD, Mena LJ. Computer Simulation of Catheter Cryoablation for Pulmonary Vein Isolation. Healthcare (Basel) 2024; 12:1508. [PMID: 39120212 PMCID: PMC11312416 DOI: 10.3390/healthcare12151508] [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: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Cryoablation is a well-established medical procedure for surgically treating atrial fibrillation. Cryothermal catheter therapy induces cellular necrosis by freezing the insides of pulmonary veins, with the goal of disrupting abnormal electrical heart signals. Nevertheless, tissue damage induced by cold temperatures may also lead to other complications after cardiac surgery. In this sense, the simulation of catheter ablation can provide safer environments for training and the performance of cryotherapy interventions. Therefore, in this paper, we propose a novel approach to help better understand how temperature rates can affect this procedure by using computer tools to develop a simulation framework to predict lesion size and determine optimal temperature conditions for reducing the risk of major complications. The results showed that a temperature profile of around -40 °C caused less penetration, reduced necrotic damage, and smaller lesion size in the tissue. Instead, cryotherapy close to -60 °C achieved a greater depth of temperature flow inside the tissue and a larger cross-section area of the lesion. With further development and validation, the framework could represent a cost-effective strategy for providing personalized modeling, better planning of cryocatheter-based treatment, and preventing surgical complications.
Collapse
Affiliation(s)
- Solange I. Rivera
- Department of Chemical, Electronic and Biomedical Engineering, Science and Engineering Division, Universidad de Guanajuato, Guanajuato 36250, Mexico;
| | - Clara P. Bernal
- Faculty of Engineering and Technologies, Universidad La Salle Bajío, Leon 36700, Mexico;
| | - Rafael Martínez-Peláez
- Department of Systems and Computer Engineering, Faculty of Engineering and Geological Sciences, Universidad Católica del Norte, Antofagasta 1270709, Chile;
- Computer Academic Unit, School of Information Technologies, Universidad Politécnica de Sinaloa, Mazatlan 82199, Mexico; (V.G.F.); (R.O.)
| | - Rogelio Robledo-Nolasco
- Department of Interventional Cardiology and Electrophysiology, Centro Médico Nacional 20 de Noviembre ISSSTE, Mexico City 03100, Mexico; (R.R.-N.); (G.D.L.-L.)
| | - Gerardo De León-Larios
- Department of Interventional Cardiology and Electrophysiology, Centro Médico Nacional 20 de Noviembre ISSSTE, Mexico City 03100, Mexico; (R.R.-N.); (G.D.L.-L.)
| | - Vanessa G. Félix
- Computer Academic Unit, School of Information Technologies, Universidad Politécnica de Sinaloa, Mazatlan 82199, Mexico; (V.G.F.); (R.O.)
| | - Rodolfo Ostos
- Computer Academic Unit, School of Information Technologies, Universidad Politécnica de Sinaloa, Mazatlan 82199, Mexico; (V.G.F.); (R.O.)
| | - Gladys E. Maestre
- Institute of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78550, USA; (G.E.M.); (J.D.M.)
| | - Jesús D. Melgarejo
- Institute of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78550, USA; (G.E.M.); (J.D.M.)
| | - Luis J. Mena
- Computer Academic Unit, School of Information Technologies, Universidad Politécnica de Sinaloa, Mazatlan 82199, Mexico; (V.G.F.); (R.O.)
| |
Collapse
|
9
|
Gutor SS, Richmond BW, Agrawal V, Brittain EL, Shaver CM, Wu P, Boyle TK, Mallugari RR, Douglas K, Piana RN, Johnson JE, Miller RF, Newman JH, Blackwell TS, Polosukhin VV. Pulmonary vascular disease in Veterans with post-deployment respiratory syndrome. Cardiovasc Pathol 2024; 71:107640. [PMID: 38604505 DOI: 10.1016/j.carpath.2024.107640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024] Open
Abstract
Exertional dyspnea has been documented in US military personnel after deployment to Iraq and Afghanistan. We studied whether continued exertional dyspnea in this patient population is associated with pulmonary vascular disease (PVD). We performed detailed histomorphometry of pulmonary vasculature in 52 Veterans with biopsy-proven post-deployment respiratory syndrome (PDRS) and then recruited five of these same Veterans with continued exertional dyspnea to undergo a follow-up clinical evaluation, including symptom questionnaire, pulmonary function testing, surface echocardiography, and right heart catheterization (RHC). Morphometric evaluation of pulmonary arteries showed significantly increased intima and media thicknesses, along with collagen deposition (fibrosis), in Veterans with PDRS compared to non-diseased (ND) controls. In addition, pulmonary veins in PDRS showed increased intima and adventitia thicknesses with prominent collagen deposition compared to controls. Of the five Veterans involved in our clinical follow-up study, three had borderline or overt right ventricle (RV) enlargement by echocardiography and evidence of pulmonary hypertension (PH) on RHC. Together, our studies suggest that PVD with predominant venular fibrosis is common in PDRS and development of PH may explain exertional dyspnea and exercise limitation in some Veterans with PDRS.
Collapse
Affiliation(s)
- Sergey S Gutor
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Bradley W Richmond
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Veterans Affairs, Nashville VA, Nashville, TN; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Ciara M Shaver
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Pingsheng Wu
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN
| | - Taryn K Boyle
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Ravinder R Mallugari
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Katrina Douglas
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Robert N Piana
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joyce E Johnson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Robert F Miller
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - John H Newman
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Veterans Affairs, Nashville VA, Nashville, TN; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Vasiliy V Polosukhin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
10
|
Heise EL, Salman J, Webs KS, Höffler K, Brandenberger C, Böthig D, Mühlfeld C, Haverich A. Hypoxic perfusion of pulmonary arterial vasa vasorum increases pulmonary arterial pressure. Am J Physiol Lung Cell Mol Physiol 2024; 327:L79-L85. [PMID: 38651234 DOI: 10.1152/ajplung.00346.2023] [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: 11/09/2023] [Revised: 01/22/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
The pathophysiology of pulmonary hypertension (PH) is not fully understood. Here, we tested the hypothesis that hypoxic perfusion of the vasa vasorum of the pulmonary arterial (PA) wall causes PH. Young adult pig lungs were explanted and placed into a modified ex vivo lung perfusion unit (organ care system, OCS) allowing the separate adjustment of parameters for mechanical ventilation, as well as PA perfusion and bronchial arterial (BA) perfusion. The PA vasa vasorum are branches of the BA. The lungs were used either as the control group (n = 3) or the intervention group (n = 8). The protocol for the intervention group was as follows: normoxic ventilation and perfusion (steady state), hypoxic BA perfusion, steady state, and hypoxic BA perfusion. During hypoxic BA perfusion, ventilation and PA perfusion maintained normal. Control lungs were kept under steady-state conditions for 105 min. During the experiments, PA pressure (PAP) and blood gas analysis were frequently monitored. Hypoxic perfusion of the BA resulted in an increase in systolic and mean PAP, a reaction that was reversible upon normoxic BA perfusion. The PAP increase was reproducible during the second hypoxic BA perfusion. Under control conditions, the PAP stayed constant until about 80 min of the experiment. In conclusion, the results of the current study prove that hypoxic perfusion of the vasa vasorum of the PA directly increases PAP in an ex situ lung perfusion setup, suggesting that PA vasa vasorum function and wall ischemia may contribute to the development of PH.NEW & NOTEWORTHY Hypoxic perfusion of the vasa vasorum of the pulmonary artery directly increased pulmonary arterial pressure in an ex vivo lung perfusion setup. This suggests that the function of pulmonary arterial vasa vasorum and wall ischemia may contribute to the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Emma L Heise
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Jawad Salman
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Karolin S Webs
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Klaus Höffler
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Christina Brandenberger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Hannover Medical School, Institute of Functional and Applied Anatomy, Hannover, Germany
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Böthig
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Christian Mühlfeld
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Hannover Medical School, Institute of Functional and Applied Anatomy, Hannover, Germany
| | - Axel Haverich
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| |
Collapse
|
11
|
Raslan AA, Pham TX, Lee J, Kontodimas K, Tilston-Lunel A, Schmottlach J, Hong J, Dinc T, Bujor AM, Caporarello N, Thiriot A, von Andrian UH, Huang SK, Nicosia RF, Trojanowska M, Varelas X, Ligresti G. Lung injury-induced activated endothelial cell states persist in aging-associated progressive fibrosis. Nat Commun 2024; 15:5449. [PMID: 38937456 PMCID: PMC11211333 DOI: 10.1038/s41467-024-49545-x] [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: 02/08/2023] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
Progressive lung fibrosis is associated with poorly understood aging-related endothelial cell dysfunction. To gain insight into endothelial cell alterations in lung fibrosis we performed single cell RNA-sequencing of bleomycin-injured lungs from young and aged mice. Analysis reveals activated cell states enriched for hypoxia, glycolysis and YAP/TAZ activity in ACKR1+ venous and TrkB+ capillary endothelial cells. Endothelial cell activation is prevalent in lungs of aged mice and can also be detected in human fibrotic lungs. Longitudinal single cell RNA-sequencing combined with lineage tracing demonstrate that endothelial activation resolves in young mouse lungs but persists in aged ones, indicating a failure of the aged vasculature to return to quiescence. Genes associated with activated lung endothelial cells states in vivo can be induced in vitro by activating YAP/TAZ. YAP/TAZ also cooperate with BDNF, a TrkB ligand that is reduced in fibrotic lungs, to promote capillary morphogenesis. These findings offer insights into aging-related lung endothelial cell dysfunction that may contribute to defective lung injury repair and persistent fibrosis.
Collapse
Affiliation(s)
- Ahmed A Raslan
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Tho X Pham
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jillian Schmottlach
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jeongmin Hong
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Taha Dinc
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andreea M Bujor
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | | | - Aude Thiriot
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Trojanowska
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| | - Giovanni Ligresti
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
12
|
Horvat D, Agoston-Coldea L. A spotlight on the aged pulmonary artery. Adv Clin Chem 2024; 123:157-177. [PMID: 39181621 DOI: 10.1016/bs.acc.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The ever-increasing life expectancy of the global population introduces a critical perspective on the impact of aging as an immutable cardiovascular risk factor, particularly manifesting in the alterations observed in the pulmonary artery (PA). Mechanisms contributing to aging-induced changes in PA include endothelial dysfunction, chronic inflammation, and structural changes in the arterial wall over time. These alterations extend beyond mere elasticity, exerting profound effects on pulmonary hemodynamics. The propensity of PAs to develop atherosclerotic plaques underscores an intriguing facet of vascular aging, although the available literature is currently insufficient to comprehensively assess their true incidence. While recognizing the inherent risk of periprocedural complications, right heart catheterization (RHC) stands out as the gold standard for precise hemodynamic evaluation. Echocardiography, a widely employed method, proves valuable for screening pulmonary hypertension (PH), yet falls short of diagnostic capability. Technological advancements usher in a new era with non-invasive modalities such as cardiac magnetic resonance (CMR) imaging emerging as promising tools. These innovations demonstrate their prowess in providing accurate assessments of PA stiffness and hemodynamics, offering a glimpse into the future landscape of diagnostic methodologies. As we navigate the intersection of aging and pulmonary vascular health, this review aims to address mechanisms and techniques for assessing PA aging, highlighting the need for comprehensive assessments to guide clinical decision making in an increasingly aging population.
Collapse
Affiliation(s)
- Dalma Horvat
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; 2nd Department of Internal Medicine, Emergency County Hospital, Cluj-Napoca, Romania.
| |
Collapse
|
13
|
Sinitca AM, Lyanova AI, Kaplun DI, Hassan H, Krasichkov AS, Sanarova KE, Shilenko LA, Sidorova EE, Akhmetova AA, Vaulina DD, Karpov AA. Microscopy Image Dataset for Deep Learning-Based Quantitative Assessment of Pulmonary Vascular Changes. Sci Data 2024; 11:635. [PMID: 38879569 PMCID: PMC11180164 DOI: 10.1038/s41597-024-03473-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/04/2024] [Indexed: 06/19/2024] Open
Abstract
Pulmonary hypertension (PH) is a syndrome complex that accompanies a number of diseases of different etiologies, associated with basic mechanisms of structural and functional changes of the pulmonary circulation vessels and revealed pressure increasing in the pulmonary artery. The structural changes in the pulmonary circulation vessels are the main limiting factor determining the prognosis of patients with PH. Thickening and irreversible deposition of collagen in the pulmonary artery branches walls leads to rapid disease progression and a therapy effectiveness decreasing. In this regard, histological examination of the pulmonary circulation vessels is critical both in preclinical studies and clinical practice. However, measurements of quantitative parameters such as the average vessel outer diameter, the vessel walls area, and the hypertrophy index claimed significant time investment and the requirement for specialist training to analyze micrographs. A dataset of pulmonary circulation vessels for pathology assessment using semantic segmentation techniques based on deep-learning is presented in this work. 609 original microphotographs of vessels, numerical data from experts' measurements, and microphotographs with outlines of these measurements for each of the vessels are presented. Furthermore, here we cite an example of a deep learning pipeline using the U-Net semantic segmentation model to extract vascular regions. The presented database will be useful for the development of new software solutions for the analysis of histological micrograph.
Collapse
Affiliation(s)
- Aleksandr M Sinitca
- Centre for Digital Telecommunication Technologies, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
| | - Asya I Lyanova
- Centre for Digital Telecommunication Technologies, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
| | - Dmitrii I Kaplun
- Artificial Intelligence Research Institute, China University of Mining and Technology, Xuzhou, 221116, China.
- Department of Automation and Control Processes, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia.
| | - Hassan Hassan
- Department of Automation and Control Processes, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
| | - Alexander S Krasichkov
- Radio Engineering Systems Department, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
- Department of Computer Science and Engineering, St. Petersburg Electrotechnical University "LETI", 197022, Saint Petersburg, Russia
| | - Kseniia E Sanarova
- Radio Engineering Systems Department, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
| | - Leonid A Shilenko
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, 197341, Russia
| | - Elizaveta E Sidorova
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, 197341, Russia
| | - Anna A Akhmetova
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, 197341, Russia
| | - Dariya D Vaulina
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, 197341, Russia
| | - Andrei A Karpov
- Department of Computer Science and Engineering, St. Petersburg Electrotechnical University "LETI", 197022, Saint Petersburg, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, 197341, Russia
| |
Collapse
|
14
|
Esposito TVF, Blackadar C, Wu L, Rodríguez-Rodríguez C, Haney EF, Pletzer D, Saatchi K, Hancock REW, Häfeli UO. Biodistribution of Native and Nanoformulated Innate Defense Regulator Peptide 1002. Mol Pharm 2024; 21:2751-2766. [PMID: 38693707 DOI: 10.1021/acs.molpharmaceut.3c01169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Innate defense regulator-1002 (IDR-1002) is a synthetic peptide with promising immunomodulatory and antibiofilm properties. An appreciable body of work exists around its mechanism of action at the cellular and molecular level, along with its efficacy across several infection and inflammation models. However, little is known about its absorption, distribution, and excretion in live organisms. Here, we performed a comprehensive biodistribution assessment with a gallium-67 radiolabeled derivative of IDR-1002 using nuclear tracing techniques. Various dose levels of the radiotracer (2-40 mg/kg) were administered into the blood, peritoneal cavity, and subcutaneous tissue, or instilled into the lungs. The peptide was well tolerated at all subcutaneous and intraperitoneal doses, although higher levels were associated with delayed absorption kinetics and precipitation of the peptide within the tissues. Low intratracheal doses were rapidly absorbed systemically, and small increases in the dose level were lethal. Intravenous doses were rapidly cleared from the blood at lower levels, and upon escalation, were toxic with a high proportion of the dose accumulating within the lung tissue. To improve biocompatibility and prolong its circulation within the blood, IDR-1002 was further formulated onto high molecular weight hyperbranched polyglycerol (HPG) polymers. Constructs prepared at 5:1 and 10:1 peptide-to-polymer ratios were colloidally stable, maintained the biological profile of the peptide payload and helped reduce red blood cell lysis. The 5:1 construct circulated well in the blood, but higher peptide loading was associated with rapid clearance by the reticuloendothelial system. Many peptides face pharmacokinetic and biocompatibility challenges, but formulations such as those with HPG have the potential to overcome these limitations.
Collapse
Affiliation(s)
- Tullio V F Esposito
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Colin Blackadar
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Lan Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, Shenyang 110016, China
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
- Department of Physics and Astronomy, Faculty of Science, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Evan F Haney
- Centre for Microbial Disease and Immunity Research, Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
- Asep Medical Holdings, 420 - 730 View Street, Victoria V8W 3Y7, British Columbia, Canada
| | - Daniel Pletzer
- Centre for Microbial Disease and Immunity Research, Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Robert E W Hancock
- Centre for Microbial Disease and Immunity Research, Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 1172, Denmark
| |
Collapse
|
15
|
Manning EP, Mishall P, Ramachandra AB, Hassab AHM, Lamy J, Peters DC, Murphy TE, Heerdt P, Singh I, Downie S, Choudhary G, Tellides G, Humphrey JD. Stiffening of the human proximal pulmonary artery with increasing age. Physiol Rep 2024; 12:e16090. [PMID: 38884325 PMCID: PMC11181131 DOI: 10.14814/phy2.16090] [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: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Adverse effects of large artery stiffening are well established in the systemic circulation; stiffening of the proximal pulmonary artery (PPA) and its sequelae are poorly understood. We combined in vivo (n = 6) with ex vivo data from cadavers (n = 8) and organ donors (n = 13), ages 18 to 89, to assess whether aging of the PPA associates with changes in distensibility, biaxial wall strain, wall thickness, vessel diameter, and wall composition. Aging exhibited significant negative associations with distensibility and cyclic biaxial strain of the PPA (p ≤ 0.05), with decreasing circumferential and axial strains of 20% and 7%, respectively, for every 10 years after 50. Distensibility associated directly with diffusion capacity of the lung (R2 = 0.71, p = 0.03). Axial strain associated with right ventricular ejection fraction (R2 = 0.76, p = 0.02). Aging positively associated with length of the PPA (p = 0.004) and increased luminal caliber (p = 0.05) but showed no significant association with mean wall thickness (1.19 mm, p = 0.61) and no significant differences in the proportions of mural elastin and collagen (p = 0.19) between younger (<50 years) and older (>50) ex vivo samples. We conclude that age-related stiffening of the PPA differs from that of the aorta; microstructural remodeling, rather than changes in overall geometry, may explain age-related stiffening.
Collapse
Affiliation(s)
- Edward P. Manning
- Section of Pulmonary, Critical Care, and Pulmonary MedicineYale School of MedicineNew HavenConnecticutUSA
- VA Connecticut Healthcare SystemWest HavenConnecticutUSA
| | - Priti Mishall
- Department of Anatomy and Structural BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of Ophthalmology and Visual SciencesAlbert Einstein College of MedicineBronxNew YorkUSA
| | | | | | - Jerome Lamy
- Université Paris Cité, INSERM U970, PARCC, APHP Hôpital Européen Georges PompidouParisFrance
| | - Dana C. Peters
- Department of RadiologyYale School of MedicineNew HavenConnecticutUSA
| | - Terrence E. Murphy
- Department of Public Health SciencesThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Paul Heerdt
- Department of AnesthesiologyYale School of MedicineNew HavenConnecticutUSA
| | - Inderjit Singh
- Section of Pulmonary, Critical Care, and Pulmonary MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Sherry Downie
- Department of Anatomy and Structural BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Gaurav Choudhary
- Lifespan Cardiovascular Institute, Providence VA Medical CenterProvidenceRhode IslandUSA
- Warren Alpert Medical School, Brown UniversityProvidenceRhode IslandUSA
| | - George Tellides
- VA Connecticut Healthcare SystemWest HavenConnecticutUSA
- Department of Surgery (Cardiac)Yale School of MedicineNew HavenConnecticutUSA
| | - Jay D. Humphrey
- Department of Biomedical EngineeringYale UniversityNew HavenConnecticutUSA
| |
Collapse
|
16
|
Sreepadmanabh M, Arun AB, Bhattacharjee T. Design approaches for 3D cell culture and 3D bioprinting platforms. BIOPHYSICS REVIEWS 2024; 5:021304. [PMID: 38765221 PMCID: PMC11101206 DOI: 10.1063/5.0188268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/01/2024] [Indexed: 05/21/2024]
Abstract
The natural habitat of most cells consists of complex and disordered 3D microenvironments with spatiotemporally dynamic material properties. However, prevalent methods of in vitro culture study cells under poorly biomimetic 2D confinement or homogeneous conditions that often neglect critical topographical cues and mechanical stimuli. It has also become increasingly apparent that cells in a 3D conformation exhibit dramatically altered morphological and phenotypical states. In response, efforts toward designing biomaterial platforms for 3D cell culture have taken centerstage over the past few decades. Herein, we present a broad overview of biomaterials for 3D cell culture and 3D bioprinting, spanning both monolithic and granular systems. We first critically evaluate conventional monolithic hydrogel networks, with an emphasis on specific experimental requirements. Building on this, we document the recent emergence of microgel-based 3D growth media as a promising biomaterial platform enabling interrogation of cells within porous and granular scaffolds. We also explore how jammed microgel systems have been leveraged to spatially design and manipulate cellular structures using 3D bioprinting. The advent of these techniques heralds an unprecedented ability to experimentally model complex physiological niches, with important implications for tissue bioengineering and biomedical applications.
Collapse
Affiliation(s)
- M Sreepadmanabh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| | - Ashitha B. Arun
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| | - Tapomoy Bhattacharjee
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| |
Collapse
|
17
|
Koike H, Sueyoshi E, Somagawa C, Nagayama H, Toya R. CT findings, prognosis, and follow-up of pulmonary artery periadventitial hematoma with aortic dissection: a retrospective single-center study. J Thorac Dis 2024; 16:3031-3041. [PMID: 38883683 PMCID: PMC11170392 DOI: 10.21037/jtd-23-1914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/01/2024] [Indexed: 06/18/2024]
Abstract
Background Pulmonary artery periadventitial hematoma (PAPH) with aortic dissection (AD) is a rare condition but has been reported to correlate with prognosis. However, there are few cases of PAPH, and the relationship with computed tomography (CT) findings of AD are unknown. This study aimed to evaluate CT findings and early prognosis in patients with PAPH in AD. Methods This was a retrospective analysis of data from patients with Stanford type A AD diagnosed with contrast-enhanced CT in our institution from April 2008 to February 2023; 316 patients were included in the analyses. Patients comprised a PAPH group (n=78) and a non-PAPH group (n=238). The PAPH group was further divided into a group that died within 1 week of onset (death group; n=15) and a group that survived (alive group; n=63). PAPH was classified into three grades on the basis of the CT findings, as follows: Grade 1: PAPH only in the mediastinum; Grade 2: PAPH that extended into the lung field, with/without interlobular septa; and Grade 3: PAPH with pulmonary hemorrhage. Results Compared with the non-PAPH group, the PAPH group had higher rates of early death (P=0.001), pericardial (P<0.001) and mediastinal hemorrhage (P<0.001). When comparing the death and alive groups, there was a significant difference in the rates of inoperable case (P<0.001), Grade 3 PAPH (PAPH with pulmonary hemorrhage) (P<0.001), and hemothorax (P=0.02). Multivariable analysis showed a significant association between Grade 3 PAPH (PAPH with pulmonary hemorrhage) and early death (P=0.004). Conclusions Standard type A AD with PAPH is not rare. Mortality was higher in the PAPH group vs. the non-PAPH group, and Grade 3 PAPH (PAPH with pulmonary hemorrhage) was a significant risk factor for early death.
Collapse
Affiliation(s)
- Hirofumi Koike
- Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Eijun Sueyoshi
- Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Chika Somagawa
- Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Hiroki Nagayama
- Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Ryo Toya
- Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| |
Collapse
|
18
|
Bartololo MA, Taylor-LaPole AM, Gandhi D, Johnson A, Li Y, Slack E, Stevens I, Turner Z, Weigand JD, Puelz C, Husmeier D, Olufsen MS. Computational framework for the generation of one-dimensional vascular models accounting for uncertainty in networks extracted from medical images. ARXIV 2024:arXiv:2309.08779v3. [PMID: 38313199 PMCID: PMC10836077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
One-dimensional (1D) cardiovascular models offer a non-invasive method to answer medical questions, including predictions of wave-reflection, shear stress, functional flow reserve, vascular resistance, and compliance. This model type can predict patient-specific outcomes by solving 1D fluid dynamics equations in geometric networks extracted from medical images. However, the inherent uncertainty in in-vivo imaging introduces variability in network size and vessel dimensions, affecting hemodynamic predictions. Understanding the influence of variation in image-derived properties is essential to assess the fidelity of model predictions. Numerous programs exist to render three-dimensional surfaces and construct vessel centerlines. Still, there is no exact way to generate vascular trees from the centerlines while accounting for uncertainty in data. This study introduces an innovative framework employing statistical change point analysis to generate labeled trees that encode vessel dimensions and their associated uncertainty from medical images. To test this framework, we explore the impact of uncertainty in 1D hemodynamic predictions in a systemic and pulmonary arterial network. Simulations explore hemodynamic variations resulting from changes in vessel dimensions and segmentation; the latter is achieved by analyzing multiple segmentations of the same images. Results demonstrate the importance of accurately defining vessel radii and lengths when generating high-fidelity patient-specific hemodynamics models.
Collapse
Affiliation(s)
- Michelle A Bartololo
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| | - Alyssa M Taylor-LaPole
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| | - Darsh Gandhi
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
- Department of Mathematics, University of Texas at Arlington, Arlington, TX, USA
| | - Alexandria Johnson
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
- Department of Mathematics and Statistics, University of South Florida, Tampa, FL, USA
| | - Yaqi Li
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
- North Carolina School of Science and Mathematics, Durham, NC, USA
| | - Emma Slack
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
- Department of Mathematics, Colorado State University, Fort Collins, CO, USA
| | - Isaiah Stevens
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| | - Zachary Turner
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, USA
| | - Justin D Weigand
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Charles Puelz
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Dirk Husmeier
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Mette S Olufsen
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
19
|
Criscuolo ER, Fu Y, Hao Y, Zhang Z, Yang D. A comprehensive lung CT landmark pair dataset for evaluating deformable image registration algorithms. Med Phys 2024; 51:3806-3817. [PMID: 38478966 PMCID: PMC11302745 DOI: 10.1002/mp.17026] [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: 09/19/2023] [Revised: 01/29/2024] [Accepted: 03/03/2024] [Indexed: 05/08/2024] Open
Abstract
PURPOSE Deformable image registration (DIR) is a key enabling technology in many diagnostic and therapeutic tasks, but often does not meet the required robustness and accuracy for supporting clinical tasks. This is in large part due to a lack of high-quality benchmark datasets by which new DIR algorithms can be evaluated. Our team was supported by the National Institute of Biomedical Imaging and Bioengineering to develop DIR benchmark dataset libraries for multiple anatomical sites, comprising of large numbers of highly accurate landmark pairs on matching blood vessel bifurcations. Here we introduce our lung CT DIR benchmark dataset library, which was developed to improve upon the number and distribution of landmark pairs in current public lung CT benchmark datasets. ACQUISITION AND VALIDATION METHODS Thirty CT image pairs were acquired from several publicly available repositories as well as authors' institution with IRB approval. The data processing workflow included multiple steps: (1) The images were denoised. (2) Lungs, airways, and blood vessels were automatically segmented. (3) Bifurcations were directly detected on the skeleton of the segmented vessel tree. (4) Falsely identified bifurcations were filtered out using manually defined rules. (5) A DIR was used to project landmarks detected on the first image onto the second image of the image pair to form landmark pairs. (6) Landmark pairs were manually verified. This workflow resulted in an average of 1262 landmark pairs per image pair. Estimates of the landmark pair target registration error (TRE) using digital phantoms were 0.4 mm ± 0.3 mm. DATA FORMAT AND USAGE NOTES The data is published in Zenodo at https://doi.org/10.5281/zenodo.8200423. Instructions for use can be found at https://github.com/deshanyang/Lung-DIR-QA. POTENTIAL APPLICATIONS The dataset library generated in this work is the largest of its kind to date and will provide researchers with a new and improved set of ground truth benchmarks for quantitatively validating DIR algorithms within the lung.
Collapse
Affiliation(s)
| | - Yabo Fu
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yao Hao
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zhendong Zhang
- Department of Radiation Oncology, Duke University, Durham, NC, 27701, USA
| | - Deshan Yang
- Department of Radiation Oncology, Duke University, Durham, NC, 27701, USA
| |
Collapse
|
20
|
Saggu M, Bou-Assaf GM, Bucher R, Budyak IL, Buecheler J, Cao S, Groenning M, Khan E, Kwok SC, Minocha S, Pisupati K, Radhakrishnan V, Videbæk N, Yang DT, Zhang B, Narhi LO. Evaluating Clinical Safety and Analytical Impact of Subvisible Silicone Oil Particles in Biopharmaceutical Products. J Pharm Sci 2024; 113:1401-1414. [PMID: 38220088 DOI: 10.1016/j.xphs.2024.01.002] [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: 09/25/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
Silicone oil is a commonly used lubricant in pre-filled syringes (PFSs) and can migrate over time into solution in the form of silicone oil particles (SiOPs). The presence of these SiOPs can result in elevated subvisible particle counts in PFS drug products compared to other drug presentations such as vials or cartridges. Their presence in products presents analytical challenges as they complicate quantitation and characterization of other types of subvisible particles in solution. Previous studies have suggested that they can potentially act as adjuvant resulting in potential safety risks for patients. In this paper we present several analytical case studies describing the impact of the presence of SiOPs in biotherapeutics on the analysis of the drug as well as clinical case studies examining the effect of SiOPs on patient safety. The analytical case studies demonstrate that orthogonal techniques, especially flow imaging, can help differentiate SiOPs from other types of particulate matter. The clinical case studies showed no difference in the observed patient safety profile across multiple drugs, patient populations, and routes of administration, indicating that the presence of SiOPs does not impact patient safety.
Collapse
Affiliation(s)
- Miguel Saggu
- Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | | | - Reinhard Bucher
- Product Development Safety, Clinical Safety, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ivan L Budyak
- Biopharmaceutical Research and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
| | - Jakob Buecheler
- Technical Research and Development, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Shawn Cao
- Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Minna Groenning
- CMC Biophysical Analysis, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Emon Khan
- Late Respiratory and Immunology, AstraZeneca, Academy House, Cambridge, UK
| | - Stanley C Kwok
- Dosage Form Design & Development, BioPharmaceuticals Development, AstraZeneca, One Medimmune Way, Gaithersburg, Maryland 20878, USA
| | - Shalini Minocha
- Injectable Drug Product Development, Alexion, AstraZeneca Rare Disease, 121 Seaport Blvd, Boston, Massachusetts, USA
| | - Karthik Pisupati
- Biopharmaceutical Research and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
| | - Vinay Radhakrishnan
- Injectable Drug Product Development, Alexion, AstraZeneca Rare Disease, 121 Seaport Blvd, Boston, Massachusetts, USA
| | - Nicoline Videbæk
- Clinical Immunogenicity Analysis, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Dennis T Yang
- Biopharmaceutical Research and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
| | - Bin Zhang
- Injectable Drug Product Development, Alexion, AstraZeneca Rare Disease, 121 Seaport Blvd, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Angelini M, Belletti A, Landoni G, Zangrillo A, De Cobelli F, Palumbo D. Macklin Effect: From Pathophysiology to Clinical Implication. J Cardiothorac Vasc Anesth 2024; 38:881-883. [PMID: 38378321 DOI: 10.1053/j.jvca.2023.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/30/2023] [Accepted: 12/16/2023] [Indexed: 02/22/2024]
Abstract
Air leak syndromes (such as pneumomediastinum, pneumothorax, or subcutaneous emphysema) are frequent complications of acute respiratory distress syndrome (ARDS). Unfortunately, the development of air leaks is associated with worse outcomes. In addition, it has been hypothesized that the development of pneumomediastinum could be a marker of disease severity in patients with respiratory failure receiving noninvasive respiratory support or assisted ventilation. The so-called Macklin effect (or pulmonary interstitial emphysema) is the air dissection of the lung bronchovascular tree from peripheral to central airways following injury to distal alveoli. Ultimately, the progression of the Macklin effect leads to the development of pneumomediastinum, subcutaneous emphysema, or pneumothorax. The Macklin effect is identifiable on a chest computed tomography (CT) scan. The Macklin effect could be an accurate predictor of barotrauma in patients with ARDS (sensitivity = 89.2% [95% CI: 74.6-96.9]; specificity = 95.6% [95% CI: 90.6-98.4]), and may be a marker of disease severity. Accordingly, the detection of the Macklin effect on a chest CT scan could be used to select which patients with ARDS might benefit from different treatment algorithms, including advanced respiratory monitoring, early intubation, or, potentially, the institution of early extracorporeal support with or without invasive ventilation. In this video, the authors summarize the pathophysiology and potential clinical significance and applications of the Macklin effect in patients with acute respiratory failure.
Collapse
Affiliation(s)
- Matteo Angelini
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco De Cobelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diego Palumbo
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Wang J, Chen J, Shu L, Zhang R, Dai M, Fang X, Hu Z, Xiao L, Xi Z, Zhang J, Bao M. Carotid Baroreceptor Stimulation Improves Pulmonary Arterial Remodeling and Right Ventricular Dysfunction in Pulmonary Arterial Hypertension. JACC Basic Transl Sci 2024; 9:475-492. [PMID: 38680958 PMCID: PMC11055206 DOI: 10.1016/j.jacbts.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 05/01/2024]
Abstract
Autonomic nervous system imbalance is intricately associated with the severity and prognosis of pulmonary arterial hypertension (PAH). Carotid baroreceptor stimulation (CBS) is a nonpharmaceutical intervention for autonomic neuromodulation. The effects of CBS on monocrotaline-induced PAH were investigated in this study, and its underlying mechanisms were elucidated. The results indicated that CBS improved pulmonary hemodynamic status and alleviated right ventricular dysfunction, improving pulmonary arterial remodeling and right ventricular remodeling, thus enhancing the survival rate of monocrotaline-induced PAH rats. The beneficial effects of CBS treatment on PAH might be mediated through the inhibition of sympathetic overactivation and inflammatory immune signaling pathways.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling Shu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ruoliu Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xuesheng Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhiling Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lingling Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhaoqing Xi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junxia Zhang
- Department of Endocrinology, Taikang Tongji (Wuhan) Hospital, Wuhan, China
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
23
|
Péchoux C, Antigny F, Perros F. A correlated light and electron microscopy approach to study the subcellular localization of phosphorylated vimentin in human lung tissue. Methods Cell Biol 2024; 187:117-137. [PMID: 38705622 DOI: 10.1016/bs.mcb.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Correlative microscopy is an important approach for bridging the resolution gap between fluorescence light and electron microscopy. Here, we describe a fast and simple method for correlative immunofluorescence and immunogold labeling on the same section to elucidate the localization of phosphorylated vimentin (P-Vim), a robust feature of pulmonary vascular remodeling in cells of human lung small arteries. The lung is a complex, soft and difficult tissue to prepare for transmission electron microscopy (TEM). Detailing the molecular composition of small pulmonary arteries (<500μm) would be of great significance for research and diagnostics. Using the classical methods of immunochemistry (either hydrophilic resin or thin cryosections), is difficult to locate small arteries for analysis by TEM. To address this problem and to observe the same structures by both light and electron microscopy, correlative microscopy is a reliable approach. Immunofluorescence enables us to know the distribution of P-Vim in cells but does not provide ultrastructural detail on its localization. Labeled structures selected by fluorescence microscope can be identified and further analyzed by TEM at high resolution. With our method, the morphology of the arteries is well preserved, enabling the localization of P-Vim inside pulmonary endothelial cells. By applying this approach, fluorescent signals can be directly correlated to the corresponding subcellular structures in areas of interest.
Collapse
Affiliation(s)
- Christine Péchoux
- Université Paris-Saclay, INRAE, AgroparisTech, GABI, Jouy-en-Josas, France; MIMA2 Imaging Core Facility, Microscopie et Imagerie des Microorganismes, Animaux et Aliments, INRAE, Jouy-en-Josas, France.
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 "Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique," Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Frédéric Perros
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon 1, Bron, France
| |
Collapse
|
24
|
Zanini F, Che X, Suresh NE, Knutsen C, Klavina P, Xie Y, Domingo-Gonzalez R, Liu M, Kum A, Jones RC, Quake SR, Alvira CM, Cornfield DN. Hyperoxia prevents the dynamic neonatal increases in lung mesenchymal cell diversity. Sci Rep 2024; 14:2033. [PMID: 38263350 PMCID: PMC10805790 DOI: 10.1038/s41598-023-50717-w] [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: 05/05/2023] [Accepted: 12/23/2023] [Indexed: 01/25/2024] Open
Abstract
Rapid expansion of the pulmonary microvasculature through angiogenesis drives alveolarization, the final stage of lung development that occurs postnatally and dramatically increases lung gas-exchange surface area. Disruption of pulmonary angiogenesis induces long-term structural and physiologic lung abnormalities, including bronchopulmonary dysplasia, a disease characterized by compromised alveolarization. Although endothelial cells are primary determinants of pulmonary angiogenesis, mesenchymal cells (MC) play a critical and dual role in angiogenesis and alveolarization. Therefore, we performed single cell transcriptomics and in-situ imaging of the developing lung to profile mesenchymal cells during alveolarization and in the context of lung injury. Specific mesenchymal cell subtypes were present at birth with increasing diversity during alveolarization even while expressing a distinct transcriptomic profile from more mature correlates. Hyperoxia arrested the transcriptomic progression of the MC, revealed differential cell subtype vulnerability with pericytes and myofibroblasts most affected, altered cell to cell communication, and led to the emergence of Acta1 expressing cells. These insights hold the promise of targeted treatment for neonatal lung disease, which remains a major cause of infant morbidity and mortality across the world.
Collapse
Affiliation(s)
- Fabio Zanini
- School of Clinical Medicine, University of New South Wales, Sydney, Australia.
- Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW, Australia.
- Evolution & Ecology Research Centre, University of New South Wales, Sydney, NSW, Australia.
| | - Xibing Che
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nina E Suresh
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Carsten Knutsen
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Paula Klavina
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Yike Xie
- School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Racquel Domingo-Gonzalez
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Min Liu
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Kum
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Jones
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Cristina M Alvira
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David N Cornfield
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
25
|
Diwan R, Bhatt HN, Beaven E, Nurunnabi M. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204:115147. [PMID: 38065244 PMCID: PMC10787600 DOI: 10.1016/j.addr.2023.115147] [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: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, and life-threatening interstitial lung disease which causes scarring in the lung parenchyma and thereby affects architecture and functioning of lung. It is an irreversible damage to lung functioning which is related to epithelial cell injury, immense accumulation of immune cells and inflammatory cytokines, and irregular recruitment of extracellular matrix. The inflammatory cytokines trigger the differentiation of fibroblasts into activated fibroblasts, also known as myofibroblasts, which further increase the production and deposition of collagen at the injury sites in the lung. Despite the significant morbidity and mortality associated with PF, there is no available treatment that efficiently and effectively treats the disease by reversing their underlying pathologies. In recent years, many therapeutic regimens, for instance, rho kinase inhibitors, Smad signaling pathway inhibitors, p38, BCL-xL/ BCL-2 and JNK pathway inhibitors, have been found to be potent and effective in treating PF, in preclinical stages. However, due to non-selectivity and non-specificity, the therapeutic molecules also result in toxicity mediated severe side effects. Hence, this review demonstrates recent advances on PF pathology, mechanism and targets related to PF, development of various drug delivery systems based on small molecules, RNAs, oligonucleotides, peptides, antibodies, exosomes, and stem cells for the treatment of PF and the progress of various therapeutic treatments in clinical trials to advance PF treatment.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States; The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
26
|
Oh SB, Cho S, Kim HJ, Kim SJ. Differential expression of the enzymes regulating myosin light chain phosphorylation are responsible for the slower relaxation of pulmonary artery than mesenteric artery in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:49-57. [PMID: 38154964 PMCID: PMC10762492 DOI: 10.4196/kjpp.2024.28.1.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/30/2023]
Abstract
While arterial tone is generally determined by the phosphorylation of Ser19 in myosin light chain (p-MLC2), Thr18/Ser19 diphosphorylation of MLC2 (pp-MLC2) has been suggested to hinder the relaxation of smooth muscle. In a dual-wire myography of rodent pulmonary artery (PA) and mesenteric artery (MA), we noticed significantly slower relaxation in PA than in MA after 80 mM KCl-induced condition (80K-contraction). Thus, we investigated the MLC2 phosphorylation and the expression levels of its regulatory enzymes; soluble guanylate cyclase (sGC), Rho-A dependent kinase (ROCK) and myosin light chain phosphatase target regulatory subunit (MYPT1). Immunoblotting showed higher sGC-α and ROCK2 in PA than MA, while sGC-β and MYPT1 levels were higher in MA than in PA. Interestingly, the level of pp-MLC2 was higher in PA than in MA without stimulation. In the 80K-contraction state, the levels of p-MLC2 and pp-MLC2 were commonly increased. Treatment with the ROCK inhibitor (Y27632, 10 μM) reversed the higher pp-MLC2 in PA. In the myography study, pharmacological inhibition of sGC (ODQ, 10 μM) slowed relaxation during washout, which was more pronounced in PA than in MA. The simultaneous treatment of Y27632 and ODQ reversed the impaired relaxation in PA and MA. Although treatment of PA with Y27632 alone could increase the rate of relaxation, it was still slower than that of MA without Y27632 treatment. Taken together, we suggest that the higher ROCK and lower MYPT in PA would have induced the higher level of MLC2 phosphorylation, which is responsible for the characteristic slow relaxation in PA.
Collapse
Affiliation(s)
- Seung Beom Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Suhan Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
27
|
Ali Z, Khan MA, Hamza A, Alzahrani AI, Alalwan N, Shabaz M, Khan F. A deep learning‐based x‐ray imaging diagnosis system for classification of tuberculosis, COVID‐19, and pneumonia traits using evolutionary algorithm. INTERNATIONAL JOURNAL OF IMAGING SYSTEMS AND TECHNOLOGY 2024; 34. [DOI: 10.1002/ima.23014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 08/25/2024]
Abstract
AbstractTo aid in detection of tuberculosis, researchers have concentrated on developing computer‐aided diagnostic technologies based on x‐ray imaging. Since it generates noninvasive standard‐of‐care data, a chest x‐ray image is one of the most often used diagnostic imaging modalities in computer‐aided solutions. Due to their significant interclass similarities and low intra‐class variation abnormalities, chest x‐ray pictures continue to pose difficulty for proper diagnosis. In this paper, a novel automated framework is proposed for the classification of tuberculosis, COVID‐19, and pneumonia from chest x‐ray images using deep learning and improved optimization technique. Two pre‐trained convolutional neural network models such as EfficientB0 and ResNet50 have been utilized and fine‐tuned based on the additional layers. Both models are trained with fixed hyperparameters on the selected datasets and obtained newly trained models. A novel feature selection technique has been proposed that selects the best features. In the novel version, distance and update position formulation has been modified. The selected features are further fused using a novel technique that is based on the serial and standard deviation threshold function. The experimental process of the proposed framework is conducted on three datasets and obtained an accuracy of 98.2%, 99.0%, and 98.7%, respectively. In addition, a detailed Wilcoxon signed‐rank analysis is conducted and shows the proposed method significance performance. Based on the results, it is concluded that the proposed method accuracy is improved after the fusion process. In addition, the comparison with recent techniques shows the proposed method as more significant in terms of accuracy and precision rate.
Collapse
Affiliation(s)
- Zeeshan Ali
- Department of Computer Science HITEC University Taxila Pakistan
| | - Muhammad Attique Khan
- Department of Computer Science HITEC University Taxila Pakistan
- Department of Computer Science and Mathematics Lebanese American University Beirut Lebanon
| | - Ameer Hamza
- Department of Computer Science HITEC University Taxila Pakistan
| | | | - Nasser Alalwan
- Computer Science Department, Community College King Saud University Riyadh Saudi Arabia
| | - Mohammad Shabaz
- Model Institute of Engineering and Technology Jammu Jammu and Kashmir India
| | - Faheem Khan
- Department of Computer Engineering Gachon University Seongnam‐si South Korea
| |
Collapse
|
28
|
Szafron JM, Yang W, Feinstein JA, Rabinovitch M, Marsden AL. A computational growth and remodeling framework for adaptive and maladaptive pulmonary arterial hemodynamics. Biomech Model Mechanobiol 2023; 22:1935-1951. [PMID: 37658985 PMCID: PMC10929588 DOI: 10.1007/s10237-023-01744-z] [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: 04/19/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023]
Abstract
Hemodynamic loading is known to contribute to the development and progression of pulmonary arterial hypertension (PAH). This loading drives changes in mechanobiological stimuli that affect cellular phenotypes and lead to pulmonary vascular remodeling. Computational models have been used to simulate mechanobiological metrics of interest, such as wall shear stress, at single time points for PAH patients. However, there is a need for new approaches that simulate disease evolution to allow for prediction of long-term outcomes. In this work, we develop a framework that models the pulmonary arterial tree through adaptive and maladaptive responses to mechanical and biological perturbations. We coupled a constrained mixture theory-based growth and remodeling framework for the vessel wall with a morphometric tree representation of the pulmonary arterial vasculature. We show that non-uniform mechanical behavior is important to establish the homeostatic state of the pulmonary arterial tree, and that hemodynamic feedback is essential for simulating disease time courses. We also employed a series of maladaptive constitutive models, such as smooth muscle hyperproliferation and stiffening, to identify critical contributors to development of PAH phenotypes. Together, these simulations demonstrate an important step toward predicting changes in metrics of clinical interest for PAH patients and simulating potential treatment approaches.
Collapse
Affiliation(s)
- Jason M Szafron
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, 94305, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Weiguang Yang
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, 94305, USA
| | - Jeffrey A Feinstein
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, 94305, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, 94305, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Alison L Marsden
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, 94305, USA.
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA.
| |
Collapse
|
29
|
Ostras O, Shponka I, Pinton G. Ultrasound imaging of lung disease and its relationship to histopathology: An experimentally validated simulation approach. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2023; 154:2410-2425. [PMID: 37850835 PMCID: PMC10586875 DOI: 10.1121/10.0021870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/19/2023]
Abstract
Lung ultrasound (LUS) is a widely used technique in clinical lung assessment, yet the relationship between LUS images and the underlying disease remains poorly understood due in part to the complexity of the wave propagation physics in complex tissue/air structures. Establishing a clear link between visual patterns in ultrasound images and underlying lung anatomy could improve the diagnostic accuracy and clinical deployment of LUS. Reverberation that occurs at the lung interface is complex, resulting in images that require interpretation of the artifacts deep in the lungs. These images are not accurate spatial representations of the anatomy due to the almost total reflectivity and high impedance mismatch between aerated lung and chest wall. Here, we develop an approach based on the first principles of wave propagation physics in highly realistic maps of the human chest wall and lung to unveil a relationship between lung disease, tissue structure, and its resulting effects on ultrasound images. It is shown that Fullwave numerical simulations of ultrasound propagation and histology-derived acoustical maps model the multiple scattering physics at the lung interface and reproduce LUS B-mode images that are comparable to clinical images. However, unlike clinical imaging, the underlying tissue structure model is known and controllable. The amount of fluid and connective tissue components in the lung were gradually modified to model disease progression, and the resulting changes in B-mode images and non-imaging reverberation measures were analyzed to explain the relationship between pathological modifications of lung tissue and observed LUS.
Collapse
Affiliation(s)
- Oleksii Ostras
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Ihor Shponka
- Department of Pathology and Forensic Medicine, Dnipro State Medical University, Dnipro, Ukraine
| | - Gianmarco Pinton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| |
Collapse
|
30
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
31
|
Korde A, Haslip M, Pednekar P, Khan A, Chioccioli M, Mehta S, Lopez-Giraldez F, Bermejo S, Rojas M, Dela Cruz C, Matthay MA, Pober JS, Pierce RW, Takyar SS. MicroRNA-1 protects the endothelium in acute lung injury. JCI Insight 2023; 8:e164816. [PMID: 37737266 PMCID: PMC10561733 DOI: 10.1172/jci.insight.164816] [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: 08/31/2022] [Accepted: 08/10/2023] [Indexed: 09/23/2023] Open
Abstract
Acute lung injury (ALI) and its most severe form, acute respiratory distress syndrome (ARDS), cause severe endothelial dysfunction in the lung, and vascular endothelial growth factor (VEGF) is elevated in ARDS. We found that the levels of a VEGF-regulated microRNA, microRNA-1 (miR-1), were reduced in the lung endothelium after acute injury. Pulmonary endothelial cell-specific (EC-specific) overexpression of miR-1 protected the lung against cell death and barrier dysfunction in both murine and human models and increased the survival of mice after pneumonia-induced ALI. miR-1 had an intrinsic protective effect in pulmonary and other types of ECs; it inhibited apoptosis and necroptosis pathways and decreased capillary leak by protecting adherens and tight junctions. Comparative gene expression analysis and RISC recruitment assays identified miR-1 targets in the context of injury, including phosphodiesterase 5A (PDE5A), angiopoietin-2 (ANGPT2), CNKSR family member 3 (CNKSR3), and TNF-α-induced protein 2 (TNFAIP2). We validated miR-1-mediated regulation of ANGPT2 in both mouse and human ECs and found that in a 119-patient pneumonia cohort, miR-1 correlated inversely with ANGPT2. These findings illustrate a previously unknown role of miR-1 as a cytoprotective orchestrator of endothelial responses to acute injury with prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Asawari Korde
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maria Haslip
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Prachi Pednekar
- Department of Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | | | - Maurizio Chioccioli
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sameet Mehta
- Department of Genetics, Yale University School Medicine, New Haven, Connecticut, USA
| | | | - Santos Bermejo
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Charles Dela Cruz
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael A. Matthay
- Cardiovascular Research Institute, Department of Medicine and Anesthesiology, UCSF, San Francisco, California, USA
| | | | | | - Shervin S. Takyar
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Sakarin S, Rungsipipat A, Surachetpong SD. Perivascular inflammatory cells and their association with pulmonary arterial remodelling in dogs with pulmonary hypertension due to myxomatous mitral valve disease. Vet Res Commun 2023; 47:1505-1521. [PMID: 36976445 DOI: 10.1007/s11259-023-10106-0] [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: 12/09/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Pulmonary hypertension (PH), an increase in pulmonary arterial pressure (PAP), may occur in dogs affected with myxomatous mitral valve disease (MMVD). Recent studies suggest that an accumulation of perivascular inflammatory cells may be involved with medial thickening which is a sign of the pulmonary artery remodelling in PH. The aim of this study was to characterise perivascular inflammatory cells in the surrounding pulmonary arteries of dogs with PH due to MMVD compared to MMVD dogs and healthy control dogs. Nineteen lung samples were collected from cadavers of small-breed dogs (control n = 5; MMVD n = 7; MMVD + PH n = 7). Toluidine blue stain and multiple IHC targeting α-SMA, vWF, CD20, CD68 and CD3 was performed to examine intimal and medial thickening, assess muscularisation of the small pulmonary arteries and characterise perivascular leucocytes. Medial thickening without intimal thickening of pulmonary arteries and muscularisation of normally non-muscularised small pulmonary arteries was observed in the MMVD and MMVD + PH groups compared with the control group. The perivascular numbers of B lymphocytes, T lymphocytes and macrophages was significantly increased in the MMVD + PH group compared with the MMVD and control groups. In contrast, the perivascular number of mast cells was significantly higher in the MMVD group compared with the MMVD + PH and control groups. This study suggested that pulmonary artery remodelling as medial thickening and muscularisation of the normally non-muscular small pulmonary arteries is accompanied by the accumulation of perivascular inflammatory cells.
Collapse
Affiliation(s)
- Siriwan Sakarin
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Anudep Rungsipipat
- Companion Animal Cancer Research Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sirilak Disatian Surachetpong
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
33
|
Grotberg JB, Romanò F. Computational pulmonary edema: A microvascular model of alveolar capillary and interstitial flow. APL Bioeng 2023; 7:036101. [PMID: 37426383 PMCID: PMC10325818 DOI: 10.1063/5.0158324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
We present a microvascular model of fluid transport in the alveolar septa related to pulmonary edema. It consists of a two-dimensional capillary sheet coursing by several alveoli. The alveolar epithelial membrane runs parallel to the capillary endothelial membrane with an interstitial layer in between, making one long septal tract. A coupled system of equations uses lubrication theory for the capillary blood, Darcy flow for the porous media of the interstitium, a passive alveolus, and the Starling equation at both membranes. Case examples include normal physiology, cardiogenic pulmonary edema, acute respiratory distress syndrome (ARDS), hypoalbuminemia, and effects of PEEP. COVID-19 has dramatically increased ARDS in the world population, raising the urgency for such a model to create an analytical framework. Under normal conditions fluid exits the alveolus, crosses the interstitium, and enters the capillary. For edema, this crossflow is reversed with fluid leaving the capillary and entering the alveolus. Because both the interstitial and capillary pressures decrease downstream, the reversal can occur within a single septal tract, with edema upstream and clearance downstream. Clinically useful solution forms are provided allowing calculation of interstitial fluid pressure, crossflows, and critical capillary pressures. Overall, the interstitial pressures are found to be significantly more positive than values used in the traditional physiological literature. That creates steep gradients near the upstream and downstream end outlets, driving significant flows toward the distant lymphatics. This new physiological flow provides an explanation to the puzzle, noted since 1896, of how pulmonary lymphatics can function so far from the alveoli: the interstitium is self-clearing.
Collapse
Affiliation(s)
- James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Francesco Romanò
- Université Lille, CNRS, ONERA, Arts et Métiers Institute of Technology, Centrale Lille, UMR 9014 LMFL-Laboratoire de Mécanique des Fluides de Lille-Kampé de Fériet, F-59000 Lille, France
| |
Collapse
|
34
|
Bai Y, Li G, Yung L, Yu PB, Ai X. Intrapulmonary arterial contraction assay reveals region-specific deregulation of vasoreactivity to lung injuries. Am J Physiol Lung Cell Mol Physiol 2023; 325:L114-L124. [PMID: 37278410 PMCID: PMC10393320 DOI: 10.1152/ajplung.00293.2022] [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: 09/12/2022] [Revised: 03/15/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023] Open
Abstract
Intrapulmonary arteries located in the proximal lung differ from those in the distal lung in size, cellular composition, and the surrounding microenvironment. However, whether these structural variations lead to region-specific regulation of vasoreactivity in homeostasis and following injury is unknown. Herein, we employ a two-step method of precision-cut lung slice (PCLS) preparation, which maintains almost intact intrapulmonary arteries, to assess contractile and relaxation responses of proximal preacinar arteries (PaAs) and distal intraacinar arteries (IaAs) in mice. We found that PaAs exhibited robust vasoconstriction in response to contractile agonists and significant nitric oxide (NO)-induced vasodilation. In comparison, IaAs were less contractile and displayed a greater relaxation response to NO. Furthermore, in a mouse model of pulmonary arterial hypertension (PAH) induced by chronic exposure to ovalbumin (OVA) allergen and hypoxia (OVA-HX), IaAs demonstrated a reduced vasocontraction despite vascular wall thickening with the emergence of new αSMA+ cells coexpressing markers of pericytes. In contrast, PaAs became hypercontractile and less responsive to NO. The reduction in relaxation of PaAs was associated with decreased expression of protein kinase G, a key component of the NO pathway, following chronic OVA-HX exposure. Taken together, the PCLS prepared using the modified preparation method enables functional evaluation of pulmonary arteries in different anatomical locations and reveals region-specific mechanisms underlying the pathophysiology of PAH in a mouse model.NEW & NOTEWORTHY Utilizing mouse precision-cut lung slices with preserved intrapulmonary vessels, we demonstrated a location-dependent structural and contractile regulation of pulmonary arteries in health and on noxious stimulations. For instance, chronic ovalbumin and hypoxic exposure increased pulmonary arterial pressure (PAH) by remodeling intraacinar arterioles to reduce vascular wall compliance while enhancing vasoconstriction in proximal preacinar arteries. These findings suggest region-specific mechanisms and therapeutic targets for pulmonary vascular diseases such as PAH.
Collapse
Affiliation(s)
- Yan Bai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Guang Li
- Department of Critical Care Medicine, Renmin Hospital and Wuhan University, Wuhan, People's Republic of China
| | - Laiming Yung
- Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Paul B Yu
- Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
35
|
Pleskova SN, Bezrukov NA, Gorshkova EN, Bobyk SZ, Lazarenko EV. Exploring the Process of Neutrophil Transendothelial Migration Using Scanning Ion-Conductance Microscopy. Cells 2023; 12:1806. [PMID: 37443839 PMCID: PMC10340179 DOI: 10.3390/cells12131806] [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: 06/16/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Abstract
The dynamics of neutrophil transendothelial migration was investigated in a model of experimental septicopyemia. Scanning ion-conductance microscopy allowed us to determine changes in morphometric characteristics of endothelial cells during this process. In the presence of a pyogenic lesion simulated by Staphylococcus aureus, such migration was accompanied by both compensatory reactions and alteration of both neutrophils and endothelial cells. Neutrophils demonstrated crawling along the contact sites between endothelial cells, swarming phenomenon, as well as anergy and formation of neutrophil extracellular traps (NETs) as a normergic state. Neutrophil swarming was accompanied by an increase in the intercellular spaces between endothelial cells. Endothelial cells decreased the area of adhesion to the substrate, which was determined by a decrease in the cell projection area, and the cell membrane was smoothed. However, endothelial cell rigidity was paradoxically unchanged compared to the control. Over time, neutrophil migration led to a more significant alteration of endothelial cells: first, shallow perforations in the membrane were formed, which were repaired rather quickly, then stress fibrils were formed, and finally, endothelial cells died and multiple perforations were formed on their membrane.
Collapse
Affiliation(s)
- Svetlana N. Pleskova
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
- Department “Nanotechnology and Biotechnology”, Nizhny Novgorod State Technical University n. a. R.E. Alekseev, 603115 Nizhny Novgorod, Russia
| | - Nikolay A. Bezrukov
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Ekaterina N. Gorshkova
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Sergey Z. Bobyk
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Ekaterina V. Lazarenko
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| |
Collapse
|
36
|
Zhao X, Alibhai D, Walsh TG, Tarassova N, Englert M, Birol SZ, Li Y, Williams CM, Neal CR, Burkard P, Cross SJ, Aitken EW, Waller AK, Beltrán JB, Gunning PW, Hardeman EC, Agbani EO, Nieswandt B, Hers I, Ghevaert C, Poole AW. Highly efficient platelet generation in lung vasculature reproduced by microfluidics. Nat Commun 2023; 14:4026. [PMID: 37419900 PMCID: PMC10329040 DOI: 10.1038/s41467-023-39598-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Platelets, small hemostatic blood cells, are derived from megakaryocytes. Both bone marrow and lung are principal sites of thrombopoiesis although underlying mechanisms remain unclear. Outside the body, however, our ability to generate large number of functional platelets is poor. Here we show that perfusion of megakaryocytes ex vivo through the mouse lung vasculature generates substantial platelet numbers, up to 3000 per megakaryocyte. Despite their large size, megakaryocytes are able repeatedly to passage through the lung vasculature, leading to enucleation and subsequent platelet generation intravascularly. Using ex vivo lung and an in vitro microfluidic chamber we determine how oxygenation, ventilation, healthy pulmonary endothelium and the microvascular structure support thrombopoiesis. We also show a critical role for the actin regulator Tropomyosin 4 in the final steps of platelet formation in lung vasculature. This work reveals the mechanisms of thrombopoiesis in lung vasculature and informs approaches to large-scale generation of platelets.
Collapse
Affiliation(s)
- Xiaojuan Zhao
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| | - Dominic Alibhai
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Tony G Walsh
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Nathalie Tarassova
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Maximilian Englert
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Semra Z Birol
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Yong Li
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Christopher M Williams
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Chris R Neal
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Philipp Burkard
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Stephen J Cross
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Elizabeth W Aitken
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Amie K Waller
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - José Ballester Beltrán
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Peter W Gunning
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Edna C Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ejaife O Agbani
- Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Bernhard Nieswandt
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Cedric Ghevaert
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Alastair W Poole
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
37
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
38
|
Gharahi H, Filonova V, Mullagura HN, Nama N, Baek S, Figueroa CA. A multiscale framework for defining homeostasis in distal vascular trees: applications to the pulmonary circulation. Biomech Model Mechanobiol 2023; 22:971-986. [PMID: 36917305 DOI: 10.1007/s10237-023-01693-7] [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: 08/10/2021] [Accepted: 01/11/2023] [Indexed: 03/16/2023]
Abstract
Pulmonary arteries constitute a low-pressure network of vessels, often characterized as a bifurcating tree with heterogeneous vessel mechanics. Understanding the vascular complexity and establishing homeostasis is important to study diseases such as pulmonary arterial hypertension (PAH). The onset and early progression of PAH can be traced to changes in the morphometry and structure of the distal vasculature. Coupling hemodynamics with vessel wall growth and remodeling (G&R) is crucial for understanding pathology at distal vasculature. Accordingly, the goal of this study is to provide a multiscale modeling framework that embeds the essential features of arterial wall constituents coupled with the hemodynamics within an arterial network characterized by an extension of Murray's law. This framework will be used to establish the homeostatic baseline characteristics of a pulmonary arterial tree, including important parameters such as vessel radius, wall thickness and shear stress. To define the vascular homeostasis and hemodynamics in the tree, we consider two timescales: a cardiac cycle and a longer period of vascular adaptations. An iterative homeostatic optimization, which integrates a metabolic cost function minimization, the stress equilibrium, and hemodynamics, is performed at the slow timescale. In the fast timescale, the pulsatile blood flow dynamics is described by a Womersley's deformable wall analytical solution. Illustrative examples for symmetric and asymmetric trees are presented that provide baseline characteristics for the normal pulmonary arterial vasculature. The results are compared with diverse literature data on morphometry, structure, and mechanics of pulmonary arteries. The developed framework demonstrates a potential for advanced parametric studies and future G&R and hemodynamics modeling of PAH.
Collapse
Affiliation(s)
- Hamidreza Gharahi
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Vasilina Filonova
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Haritha N Mullagura
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Nitesh Nama
- Department of Mechanical & Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Seungik Baek
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - C Alberto Figueroa
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
40
|
Rumph JT, Stephens VR, Ameli S, Brown LK, Rayford KJ, Nde PN, Osteen KG, Bruner-Tran KL. A Paternal Fish Oil Diet Preconception Reduces Lung Inflammation in a Toxicant-Driven Murine Model of New Bronchopulmonary Dysplasia. Mar Drugs 2023; 21:161. [PMID: 36976210 PMCID: PMC10052688 DOI: 10.3390/md21030161] [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: 12/22/2022] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
New bronchopulmonary dysplasia (BPD) is a neonatal disease that is theorized to begin in utero and manifests as reduced alveolarization due to inflammation of the lung. Risk factors for new BPD in human infants include intrauterine growth restriction (IUGR), premature birth (PTB) and formula feeding. Using a mouse model, our group recently reported that a paternal history of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increased his offspring's risk of IUGR, PTB, and new BPD. Additionally, formula supplementation of these neonates worsened the severity of pulmonary disease. In a separate study, we reported that a paternal preconception fish oil diet prevented TCDD-driven IUGR and PTB. Not surprisingly, eliminating these two major risk factors for new BPD also significantly reduced development of neonatal lung disease. However, this prior study did not examine the potential mechanism for fish oil's protective effect. Herein, we sought to determine whether a paternal preconception fish oil diet attenuated toxicant-associated lung inflammation, which is an important contributor to the pathogenesis of new BPD. Compared to offspring of standard diet TCDD-exposed males, offspring of TCDD-exposed males provided a fish oil diet prior to conception exhibited a significant reduction in pulmonary expression of multiple pro-inflammatory mediators (Tlr4, Cxcr2, Il-1 alpha). Additionally, neonatal lungs of pups born to fish oil treated fathers exhibited minimal hemorrhaging or edema. Currently, prevention of BPD is largely focused on maternal strategies to improve health (e.g., smoking cessation) or reduce risk of PTB (e.g., progesterone supplementation). Our studies in mice support a role for also targeting paternal factors to improve pregnancy outcomes and child health.
Collapse
Affiliation(s)
- Jelonia T. Rumph
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Victoria R. Stephens
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - LaKendria K. Brown
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kevin G. Osteen
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| | - Kaylon L. Bruner-Tran
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
41
|
Allen BJ, Frye H, Ramanathan R, Caggiano LR, Tabima DM, Chesler NC, Philip JL. Biomechanical and Mechanobiological Drivers of the Transition From PostCapillary Pulmonary Hypertension to Combined Pre-/PostCapillary Pulmonary Hypertension. J Am Heart Assoc 2023; 12:e028121. [PMID: 36734341 PMCID: PMC9973648 DOI: 10.1161/jaha.122.028121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Combined pre-/postcapillary pulmonary hypertension (Cpc-PH), a complication of left heart failure, is associated with higher mortality rates than isolated postcapillary pulmonary hypertension alone. Currently, knowledge gaps persist on the mechanisms responsible for the progression of isolated postcapillary pulmonary hypertension (Ipc-PH) to Cpc-PH. Here, we review the biomechanical and mechanobiological impact of left heart failure on pulmonary circulation, including mechanotransduction of these pathological forces, which lead to altered biological signaling and detrimental remodeling, driving the progression to Cpc-PH. We focus on pathologically increased cyclic stretch and decreased wall shear stress; mechanotransduction by endothelial cells, smooth muscle cells, and pulmonary arterial fibroblasts; and signaling-stimulated remodeling of the pulmonary veins, capillaries, and arteries that propel the transition from Ipc-PH to Cpc-PH. Identifying biomechanical and mechanobiological mechanisms of Cpc-PH progression may highlight potential pharmacologic avenues to prevent right heart failure and subsequent mortality.
Collapse
Affiliation(s)
- Betty J. Allen
- Department of SurgeryUniversity of Wisconsin‐MadisonMadisonWI
| | - Hailey Frye
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWI
| | - Rasika Ramanathan
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWI
| | - Laura R. Caggiano
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center and Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA
| | - Diana M. Tabima
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWI
| | - Naomi C. Chesler
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWI
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center and Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA
| | | |
Collapse
|
42
|
Zhao X, Wright A, Goertz DE. An optical and acoustic investigation of microbubble cavitation in small channels under therapeutic ultrasound conditions. ULTRASONICS SONOCHEMISTRY 2023; 93:106291. [PMID: 36640460 PMCID: PMC9852793 DOI: 10.1016/j.ultsonch.2023.106291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 06/04/2023]
Abstract
Therapeutic focused ultrasound in combination with encapsulated microbubbles is being widely investigated for its ability to elicit bioeffects in the microvasculature, such as transient permeabilization for drug delivery or at higher pressures to achieve 'antivascular' effects. While it is well established that the behaviors of microbubbles are altered when they are situated within sufficiently small vessels, there is a paucity of data examining how the bubble population dynamics and emissions change as a function of channel (vessel) diameter over a size range relevant to therapeutic ultrasound, particularly at pressures relevant to antivascular ultrasound. Here we use acoustic emissions detection and high-speed microscopy (10 kframes/s) to examine the behavior of a polydisperse clinically employed agent (Definity®) in wall-less channels as their diameters are scaled from 1200 to 15 µm. Pressures are varied from 0.1 to 3 MPa using either a 5 ms pulse or a sequence of 0.1 ms pulses spaced at 1 ms, both of which have been previously employed in an in vivo context. With increasing pressure, the 1200 µm channel - on the order of small arteries and veins - exhibited inertial cavitation, 1/2 subharmonics and 3/2 ultraharmonics, consistent with numerous previous reports. The 200 and 100 µm channels - in the size range of larger microvessels less affected by therapeutic focused ultrasound - exhibited a distinctly different behavior, having muted development of 1/2 subharmonics and 3/2 ultraharmonics and reduced persistence. These were associated with radiation forces displacing bubbles to the distal wall and inducing clusters that then rapidly dissipated along with emissions. As the diameter transitioned to 50 and then 15 µm - a size regime that is most relevant to therapeutic focused ultrasound - there was a higher threshold for the onset of inertial cavitation as well as subharmonics and ultraharmonics, which importantly had more complex orders that are not normally reported. Clusters also occurred in these channels (e.g. at 3 MPa, the mean lateral and axial sizes were 23 and 72 µm in the 15 µm channel; 50 and 90 µm in the 50 µm channel), however in this case they occupied the entire lumens and displaced the wall boundaries. Damage to the 15 µm channel was observed for both pulse types, but at a lower pressure for the long pulse. Experiments conducted with a 'nanobubble' (<0.45 µm) subpopulation of Definity followed broadly similar features to 'native' Definity, albeit at a higher pressure threshold for inertial cavitation. These results provide new insights into the behavior of microbubbles in small vessels at higher pressures and have implications for therapeutic focused ultrasound cavitation monitoring and control.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| | - Alex Wright
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - David E Goertz
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| |
Collapse
|
43
|
Goel K, Egersdorf N, Gill A, Cao D, Collum SD, Jyothula SS, Huang HJ, Sauler M, Lee PJ, Majka S, Karmouty-Quintana H, Petrache I. Characterization of pulmonary vascular remodeling and MicroRNA-126-targets in COPD-pulmonary hypertension. Respir Res 2022; 23:349. [PMID: 36522710 PMCID: PMC9756782 DOI: 10.1186/s12931-022-02267-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Despite causing increased morbidity and mortality, pulmonary hypertension (PH) in chronic obstructive pulmonary disease (COPD) patients (COPD-PH) lacks treatment, due to incomplete understanding of its pathogenesis. Hypertrophy of pulmonary arterial walls and pruning of the microvasculature with loss of capillary beds are known features of pulmonary vascular remodeling in COPD. The remodeling features of pulmonary medium- and smaller vessels in COPD-PH lungs are less well described and may be linked to maladaptation of endothelial cells to chronic cigarette smoking (CS). MicroRNA-126 (miR126), a master regulator of endothelial cell fate, has divergent functions that are vessel-size specific, supporting the survival of large vessel endothelial cells and inhibiting the proliferation of microvascular endothelial cells. Since CS decreases miR126 in microvascular lung endothelial cells, we set out to characterize the remodeling by pulmonary vascular size in COPD-PH and its relationship with miR126 in COPD and COPD-PH lungs. METHODS Deidentified lung tissue was obtained from individuals with COPD with and without PH and from non-diseased non-smokers and smokers. Pulmonary artery remodeling was assessed by ⍺-smooth muscle actin (SMA) abundance via immunohistochemistry and analyzed by pulmonary artery size. miR126 and miR126-target abundance were quantified by qPCR. The expression levels of ceramide, ADAM9, and endothelial cell marker CD31 were assessed by immunofluorescence. RESULTS Pulmonary arteries from COPD and COPD-PH lungs had significantly increased SMA abundance compared to non-COPD lungs, especially in small pulmonary arteries and the lung microvasculature. This was accompanied by significantly fewer endothelial cell markers and increased pro-apoptotic ceramide abundance. miR126 expression was significantly decreased in lungs of COPD individuals. Of the targets tested (SPRED1, VEGF, LAT1, ADAM9), lung miR126 most significantly inversely correlated with ADAM9 expression. Compared to controls, ADAM9 was significantly increased in COPD and COPD-PH lungs, predominantly in small pulmonary arteries and lung microvasculature. CONCLUSION Both COPD and COPD-PH lungs exhibited significant remodeling of the pulmonary vascular bed of small and microvascular size, suggesting these changes may occur before or independent of the clinical development of PH. Decreased miR126 expression with reciprocal increase in ADAM9 may regulate endothelial cell survival and vascular remodeling in small pulmonary arteries and lung microvasculature in COPD and COPD-PH.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
| | - Amar Gill
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Danting Cao
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center Houston, Houston, USA
| | - Soma S Jyothula
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, USA
| | - Howard J Huang
- Division of Pulmonary Critical Care, Transplant Pulmonology, Houston Methodist Hospital, Houston, USA
| | - Maor Sauler
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Yale School of Medicine , New Haven, USA
| | - Patty J Lee
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, USA
| | - Susan Majka
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA
| | - Harry Karmouty-Quintana
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, and Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA.
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA.
| |
Collapse
|
44
|
Yang GH, Kang D, An S, Ryu JY, Lee K, Kim JS, Song MY, Kim YS, Kwon SM, Jung WK, Jeong W, Jeon H. Advances in the development of tubular structures using extrusion-based 3D cell-printing technology for vascular tissue regenerative applications. Biomater Res 2022; 26:73. [PMID: 36471437 PMCID: PMC9720982 DOI: 10.1186/s40824-022-00321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/13/2022] [Indexed: 12/11/2022] Open
Abstract
Until recent, there are no ideal small diameter vascular grafts available on the market. Most of the commercialized vascular grafts are used for medium to large-sized blood vessels. As a solution, vascular tissue engineering has been introduced and shown promising outcomes. Despite these optimistic results, there are limitations to commercialization. This review will cover the need for extrusion-based 3D cell-printing technique capable of mimicking the natural structure of the blood vessel. First, we will highlight the physiological structure of the blood vessel as well as the requirements for an ideal vascular graft. Then, the essential factors of 3D cell-printing including bioink, and cell-printing system will be discussed. Afterwards, we will mention their applications in the fabrication of tissue engineered vascular grafts. Finally, conclusions and future perspectives will be discussed.
Collapse
Affiliation(s)
- Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| | - SangHyun An
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jeong Yeop Ryu
- grid.258803.40000 0001 0661 1556Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, 130 Dongdeok‑ro, Jung‑gu, Daegu, 41944 South Korea
| | - KyoungHo Lee
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Jun Sik Kim
- Preclinical Research Center, K Medi-hub, 80 Cheombok-ro, Dong-gu, Daegu, 41061 South Korea
| | - Moon-Yong Song
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Young-Sik Kim
- Medical Safety Center, Bio Division, Korea Conformity Laboratories 8, Gaetbeol-ro 145beon-gil, Yeonsu-gu, Incheon, 21999 South Korea
| | - Sang-Mo Kwon
- grid.262229.f0000 0001 0719 8572Department of Physiology, School of Medicine, Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870 South Korea
| | - Won-Kyo Jung
- grid.412576.30000 0001 0719 8994Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Daeyeon-dong, Nam-gu, Busan, 48513 South Korea
| | - Woonhyeok Jeong
- grid.412091.f0000 0001 0669 3109Department of Plastic and Reconstructive Surgery, Dongsan Medical Center, Keimyung University College of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu, 42601 South Korea
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588 South Korea
| |
Collapse
|
45
|
Grotberg JB, Romanò F. Computational pulmonary edema: A microvascular model of alveolar capillary and interstitial flow. APL Bioeng 2022; 6:046104. [PMID: 36389648 PMCID: PMC9653270 DOI: 10.1063/5.0109107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
We present a microvascular model of fluid transport in the alveolar septa related to pulmonary edema. It consists of a two-dimensional capillary sheet coursing by several alveoli. The alveolar epithelial membrane runs parallel to the capillary endothelial membrane with an interstitial layer in between, making one long septal tract. A coupled system of equations is derived using lubrication theory for the capillary blood, Darcy flow for the porous media of the interstitium, a passive alveolus, and the Starling equation at both membranes. Case examples include normal physiology, cardiogenic pulmonary edema, noncardiogenic edema Acute Respiratory Distress Syndrome (ARDS) and hypoalbuminemia, and the effects of positive end expiratory pressure. COVID-19 has dramatically increased ARDS in the world population, raising the urgency for such a model to create an analytical framework. Under normal conditions, the fluid exits the alveolus, crosses the interstitium, and enters the capillary. For edema, this crossflow is reversed with the fluid leaving the capillary and entering the alveolus. Because both the interstitial and capillary pressures decrease downstream, the reversal can occur within a single septal tract, with edema upstream and clearance downstream. Overall, the interstitial pressures are found to be significantly more positive than values used in the traditional physiological literature that creates steep gradients near the upstream and downstream end outlets, driving significant flows toward the distant lymphatics. This new physiological flow may provide a possible explanation to the puzzle, noted since 1896, of how pulmonary lymphatics can function so far from the alveoli: the interstitium can be self-clearing.
Collapse
Affiliation(s)
- James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, 1107 Gerstacker Bldg., 2200 Bonisteel Blvd., Ann Arbor, Michigan 48109-2099, USA
- Author to whom correspondence should be addressed: . Tel.: (734)-936-3834. Fax: (734)-936-1905
| | - Francesco Romanò
- Univ. Lille, CNRS, ONERA, Arts et Métiers Institute of Technology, Centrale Lille, UMR 9014, LMFL-Laboratoire de Mécanique des Fluides de Lille-Kampé de Fériet, F-59000 Lille, France
| |
Collapse
|
46
|
Zhang K, Yao E, Chuang E, Chen B, Chuang EY, Chuang PT. mTORC1 signaling facilitates differential stem cell differentiation to shape the developing murine lung and is associated with mitochondrial capacity. Nat Commun 2022; 13:7252. [PMID: 36433959 PMCID: PMC9700781 DOI: 10.1038/s41467-022-34763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
Formation of branched organs requires sequential differentiation of stem cells. In this work, we find that the conducting airways derived from SOX2+ progenitors in the murine lungs fail to form without mTOR complex 1 (mTORC1) signaling and are replaced by lung cysts. Proximal-distal patterning through transitioning of distal SOX9+ progenitors to proximal SOX2+ cells is disrupted. Mitochondria number and ATP production are reduced. Compromised mitochondrial capacity results in a similar defect as that in mTORC1-deficient lungs. This suggests that mTORC1 promotes differentiation of SOX9+ progenitors to form the conducting airways by modulating mitochondrial capacity. Surprisingly, in all mutants, saccules are produced from lung cysts at the proper developmental time despite defective branching. SOX9+ progenitors also differentiate into alveolar epithelial type I and type II cells within saccules. These findings highlight selective utilization of energy and regulatory programs during stem cell differentiation to produce distinct structures of the mammalian lungs.
Collapse
Affiliation(s)
- Kuan Zhang
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Erica Yao
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Ethan Chuang
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Biao Chen
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Evelyn Y. Chuang
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Pao-Tien Chuang
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| |
Collapse
|
47
|
Zhao T, Parmisano S, Soroureddin Z, Zhao M, Yung L, Thistlethwaite PA, Makino A, Yuan JXJ. Mechanosensitive cation currents through TRPC6 and Piezo1 channels in human pulmonary arterial endothelial cells. Am J Physiol Cell Physiol 2022; 323:C959-C973. [PMID: 35968892 PMCID: PMC9485000 DOI: 10.1152/ajpcell.00313.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022]
Abstract
Mechanosensitive cation channels and Ca2+ influx through these channels play an important role in the regulation of endothelial cell functions. Transient receptor potential canonical channel 6 (TRPC6) is a diacylglycerol-sensitive nonselective cation channel that forms receptor-operated Ca2+ channels in a variety of cell types. Piezo1 is a mechanosensitive cation channel activated by membrane stretch and shear stress in lung endothelial cells. In this study, we report that TRPC6 and Piezo1 channels both contribute to membrane stretch-mediated cation currents and Ca2+ influx or increase in cytosolic-free Ca2+ concentration ([Ca2+]cyt) in human pulmonary arterial endothelial cells (PAECs). The membrane stretch-mediated cation currents and increase in [Ca2+]cyt in human PAECs were significantly decreased by GsMTX4, a blocker of Piezo1 channels, and by BI-749327, a selective blocker of TRPC6 channels. Extracellular application of 1-oleoyl-2-acetyl-sn-glycerol (OAG), a membrane permeable analog of diacylglycerol, rapidly induced whole cell cation currents and increased [Ca2+]cyt in human PAECs and human embryonic kidney (HEK)-cells transiently transfected with the human TRPC6 gene. Furthermore, membrane stretch with hypo-osmotic or hypotonic solution enhances the cation currents in TRPC6-transfected HEK cells. In HEK cells transfected with the Piezo1 gene, however, OAG had little effect on the cation currents, but membrane stretch significantly enhanced the cation currents. These data indicate that, while both TRPC6 and Piezo1 are involved in generating mechanosensitive cation currents and increases in [Ca2+]cyt in human PAECs undergoing mechanical stimulation, only TRPC6 (but not Piezo1) is sensitive to the second messenger diacylglycerol. Selective blockers of these channels may help develop novel therapies for mechanotransduction-associated pulmonary vascular remodeling in patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Zahra Soroureddin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Manjia Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Lauren Yung
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, California
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| |
Collapse
|
48
|
Todorova B, Godon O, Conde E, Gillis CM, Iannascoli B, Richard-Le Goff O, Fiole D, Roumenina LT, Leusen JHW, Murphy AJ, Macdonald LE, Reber LL, Jönsson F, Bruhns P. IgG Subclass-Dependent Pulmonary Antigen Retention during Acute IgG-Dependent Systemic Anaphylaxis in Mice. THE JOURNAL OF IMMUNOLOGY 2022; 209:1243-1251. [DOI: 10.4049/jimmunol.2200234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/25/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Mouse models of active systemic anaphylaxis rely predominantly on IgG Abs forming IgG–allergen immune complexes that induce IgG receptor–expressing neutrophils and monocytes/macrophages to release potent mediators, leading to systemic effects. Whether anaphylaxis initiates locally or systemically remains unknown. In this study, we aimed at identifying the anatomical location of IgG–allergen immune complexes during anaphylaxis. Active systemic anaphylaxis was induced following immunization with BSA and i.v. challenge with fluorescently labeled BSA. Ag retention across different organs was examined using whole-body fluorescence imaging, comparing immunized and naive animals. Various mouse models and in vivo deletion strategies were employed to determine the contribution of IgG receptors, complement component C1q, myeloid cell types, and anaphylaxis mediators. We found that following challenge, Ag diffused systemically, but specifically accumulated in the lungs of mice sensitized to that Ag, where it formed large Ab-dependent aggregates in the vasculature. Ag retention in the lungs did not rely on IgG receptors, C1q, neutrophils, or macrophages. IgG2a-mediated, but neither IgG1- nor IgG2b-mediated, passive systemic anaphylaxis led to Ag retention in the lung. Neutrophils and monocytes significantly accumulated in the lungs after challenge and captured high amounts of Ag, which led to downmodulation of surface IgG receptors and triggered their activation. Thus, within minutes of systemic injection in sensitized mice, Ag formed aggregates in the lung and liver vasculature, but accumulated specifically and dose-dependently in the lung. Neutrophils and monocytes recruited to the lung captured Ag and became activated. However, Ag aggregation in the lung vasculature was not necessary for anaphylaxis induction.
Collapse
Affiliation(s)
- Biliana Todorova
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Ophélie Godon
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Eva Conde
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Caitlin M. Gillis
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Bruno Iannascoli
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Odile Richard-Le Goff
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Daniel Fiole
- †Unité Biothérapies Anti-Infectieuses et Immunité, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
- ‡Unit of Human Histopathology and Animal Models, Institut Pasteur, Université de Paris, Paris, France
| | - Lubka T. Roumenina
- §Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | | | | | | | - Laurent L. Reber
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
- #Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Friederike Jönsson
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| | - Pierre Bruhns
- *Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Université de Paris, INSERM UMR1222, Paris, France
| |
Collapse
|
49
|
Muhl L, Mocci G, Pietilä R, Liu J, He L, Genové G, Leptidis S, Gustafsson S, Buyandelger B, Raschperger E, Hansson EM, Björkegren JL, Vanlandewijck M, Lendahl U, Betsholtz C. A single-cell transcriptomic inventory of murine smooth muscle cells. Dev Cell 2022; 57:2426-2443.e6. [DOI: 10.1016/j.devcel.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/12/2022] [Accepted: 09/27/2022] [Indexed: 11/28/2022]
|
50
|
Ozdemir S, Yalcin-Enis I, Yalcinkaya B, Yalcinkaya F. An Investigation of the Constructional Design Components Affecting the Mechanical Response and Cellular Activity of Electrospun Vascular Grafts. MEMBRANES 2022; 12:929. [PMID: 36295688 PMCID: PMC9607146 DOI: 10.3390/membranes12100929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Cardiovascular disease is anticipated to remain the leading cause of death globally. Due to the current problems connected with using autologous arteries for bypass surgery, researchers are developing tissue-engineered vascular grafts (TEVGs). The major goal of vascular tissue engineering is to construct prostheses that closely resemble native blood vessels in terms of morphological, mechanical, and biological features so that these scaffolds can satisfy the functional requirements of the native tissue. In this setting, morphology and cellular investigation are usually prioritized, while mechanical qualities are generally addressed superficially. However, producing grafts with good mechanical properties similar to native vessels is crucial for enhancing the clinical performance of vascular grafts, exposing physiological forces, and preventing graft failure caused by intimal hyperplasia, thrombosis, aneurysm, blood leakage, and occlusion. The scaffold's design and composition play a significant role in determining its mechanical characteristics, including suturability, compliance, tensile strength, burst pressure, and blood permeability. Electrospun prostheses offer various models that can be customized to resemble the extracellular matrix. This review aims to provide a comprehensive and comparative review of recent studies on the mechanical properties of fibrous vascular grafts, emphasizing the influence of structural parameters on mechanical behavior. Additionally, this review provides an overview of permeability and cell growth in electrospun membranes for vascular grafts. This work intends to shed light on the design parameters required to maintain the mechanical stability of vascular grafts placed in the body to produce a temporary backbone and to be biodegraded when necessary, allowing an autologous vessel to take its place.
Collapse
Affiliation(s)
- Suzan Ozdemir
- Textile Engineering Department, Textile Technologies and Design Faculty, Istanbul Technical University, Beyoglu, 34467 Istanbul, Turkey
| | - Ipek Yalcin-Enis
- Textile Engineering Department, Textile Technologies and Design Faculty, Istanbul Technical University, Beyoglu, 34467 Istanbul, Turkey
| | - Baturalp Yalcinkaya
- Department of Material Science, Faculty of Mechanical Engineering, Technical University of Liberec, 461 17 Liberec, Czech Republic
| | - Fatma Yalcinkaya
- Department of Environmental Technology, Institute for Nanomaterials, Advanced Technologies and Innovations, Technical University of Liberec, 461 17 Liberec, Czech Republic
| |
Collapse
|