1
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
2
|
He J, Duan P, Liu Y, Feng T, Wang S, Lin X, Xie J, Liu X. Unveiling the Impact of Hemodynamics on Endothelial Inflammation-Mediated Hepatocellular Carcinoma Metastasis Using a Biomimetic Vascular Flow Model. Adv Healthc Mater 2024; 13:e2304439. [PMID: 38486060 DOI: 10.1002/adhm.202304439] [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: 12/13/2023] [Revised: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC) hematogenous dissemination is a leading cause of HCC-related deaths. The inflammatory facilitates this process by promoting the adhesion and invasion of tumor cells in the circulatory system. But the contribution of hemodynamics to this process remains poorly understood due to the lack of a suitable vascular flow model for investigation. This study develops a vascular flow model to examine the impact of hemodynamics on endothelial inflammation-mediated HCC metastasis. This work finds the increasing shear stress will reduce the recruitment of HCC cells by disturbing adhesion forces between endothelium and HCC cells. However, this reduction will be restored by the inflammation. When applying high FSS (4-6 dyn cm-2) to the inflammatory endothelium, there will be a 4.8-fold increase in HCC cell adhesions compared to normal condition. Nevertheless, the increase fold of cell adhesions is inapparent, around 1.5-fold, with low and medium FSS. This effect can be attributed to the FSS-induced upregulation of ICAM-1 and VCAM-1 of the inflammatory endothelium, which serve to strengthen cell binding forces. These findings indicate that hemodynamics plays a key role in HCC metastasis during endothelial inflammation by regulating the expression of adhesion-related factors.
Collapse
Affiliation(s)
- Jia He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Peiyan Duan
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yi Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuo Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xinyi Lin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Al Tabosh T, Liu H, Koça D, Al Tarrass M, Tu L, Giraud S, Delagrange L, Beaudoin M, Rivière S, Grobost V, Rondeau-Lutz M, Dupuis O, Ricard N, Tillet E, Machillot P, Salomon A, Picart C, Battail C, Dupuis-Girod S, Guignabert C, Desroches-Castan A, Bailly S. Impact of heterozygous ALK1 mutations on the transcriptomic response to BMP9 and BMP10 in endothelial cells from hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension donors. Angiogenesis 2024; 27:211-227. [PMID: 38294582 PMCID: PMC11021321 DOI: 10.1007/s10456-023-09902-8] [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: 10/04/2023] [Accepted: 12/03/2023] [Indexed: 02/01/2024]
Abstract
Heterozygous activin receptor-like kinase 1 (ALK1) mutations are associated with two vascular diseases: hereditary hemorrhagic telangiectasia (HHT) and more rarely pulmonary arterial hypertension (PAH). Here, we aimed to understand the impact of ALK1 mutations on BMP9 and BMP10 transcriptomic responses in endothelial cells. Endothelial colony-forming cells (ECFCs) and microvascular endothelial cells (HMVECs) carrying loss of function ALK1 mutations were isolated from newborn HHT and adult PAH donors, respectively. RNA-sequencing was performed on each type of cells compared to controls following an 18 h stimulation with BMP9 or BMP10. In control ECFCs, BMP9 and BMP10 stimulations induced similar transcriptomic responses with around 800 differentially expressed genes (DEGs). ALK1-mutated ECFCs unexpectedly revealed highly similar transcriptomic profiles to controls, both at the baseline and upon stimulation, and normal activation of Smad1/5 that could not be explained by a compensation in cell-surface ALK1 level. Conversely, PAH HMVECs revealed strong transcriptional dysregulations compared to controls with > 1200 DEGs at the baseline. Consequently, because our study involved two variables, ALK1 genotype and BMP stimulation, we performed two-factor differential expression analysis and identified 44 BMP9-dysregulated genes in mutated HMVECs, but none in ECFCs. Yet, the impaired regulation of at least one hit, namely lunatic fringe (LFNG), was validated by RT-qPCR in three different ALK1-mutated endothelial models. In conclusion, ALK1 heterozygosity only modified the BMP9/BMP10 regulation of few genes, including LFNG involved in NOTCH signaling. Future studies will uncover whether dysregulations in such hits are enough to promote HHT/PAH pathogenesis, making them potential therapeutic targets, or if second hits are necessary.
Collapse
Affiliation(s)
- T Al Tabosh
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - H Liu
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - D Koça
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - M Al Tarrass
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - L Tu
- Faculté de Médecine, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
| | - S Giraud
- Genetics Department, Femme-Mère-Enfants Hospital, Hospices Civils de Lyon, 69677, Bron, France
| | - L Delagrange
- Genetics Department, Femme-Mère-Enfants Hospital, Hospices Civils de Lyon, 69677, Bron, France
- National Reference Center for HHT, 69677, Bron, France
| | - M Beaudoin
- Genetics Department, Femme-Mère-Enfants Hospital, Hospices Civils de Lyon, 69677, Bron, France
- National Reference Center for HHT, 69677, Bron, France
| | - S Rivière
- Internal Medicine Department, CHU of Montpellier, St Eloi Hospital and Center of Clinical Investigation, INSERM, CIC 1411, 34295, Montpellier Cedex 7, France
| | - V Grobost
- Internal Medicine Department, CHU Estaing, 63100, Clermont-Ferrand, France
| | - M Rondeau-Lutz
- Internal Medicine Department, University Hospital of Strasbourg, 67091, Strasbourg Cedex, France
| | - O Dupuis
- Hôpital Lyon SUD, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69100, Villeurbanne, France
- Faculty of Medicine, Lyon University, 69921, Lyon, France
| | - N Ricard
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - E Tillet
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - P Machillot
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - A Salomon
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - C Picart
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - C Battail
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - S Dupuis-Girod
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
- Genetics Department, Femme-Mère-Enfants Hospital, Hospices Civils de Lyon, 69677, Bron, France
- National Reference Center for HHT, 69677, Bron, France
| | - C Guignabert
- Faculté de Médecine, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
| | - A Desroches-Castan
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France
| | - S Bailly
- Biosanté unit U1292, Grenoble Alpes University, INSERM, CEA, 38000, Grenoble, France.
| |
Collapse
|
4
|
Jing L, Liu K, Wang F, Su Y. Role of mechanically-sensitive cation channels Piezo1 and TRPV4 in trabecular meshwork cell mechanotransduction. Hum Cell 2024; 37:394-407. [PMID: 38316716 DOI: 10.1007/s13577-024-01035-4] [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: 12/05/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Glaucoma is one of the leading causes of irreversible blindness in developed countries, and intraocular pressure (IOP) is primary and only treatable risk factor, suggesting that to a significant extent, glaucoma is a disease of IOP disorder and pathological mechanotransduction. IOP-lowering ways are limited to decreaseing aqueous humour (AH) production or increasing the uveoscleral outflow pathway. Still, therapeutic approaches have been lacking to control IOP by enhancing the trabecular meshwork (TM) pathway. Trabecular meshwork cells (TMCs) have endothelial and myofibroblast properties and are responsible for the renewal of the extracellular matrix (ECM). Mechanosensitive cation channels, including Piezo1 and TRPV4, are abundantly expressed in primary TMCs and trigger mechanostress-dependent ECM and cytoskeletal remodelling. However, prolonged mechanical stimulation severely affects cellular biosynthesis through TMC mechanotransduction, including signaling, gene expression, ECM remodelling, and cytoskeletal structural changes, involving outflow facilities and elevating IOP. As for the functional coupling relationship between Piezo1 and TRPV4 channels, inspired by VECs and osteoblasts, we hypothesized that Piezo1 may also act upstream of TRPV4 in glaucomatous TM tissue, mediating the activation of TRPV4 via Ca2+ inflow or Ca2+ binding to phospholipase A2(PLA2), and thus be involved in increasing TM outflow resistance and elevated IOP. Therefore, this review aims to help identify new potential targets for IOP stabilization in ocular hypertension and primary open-angle glaucoma by understanding the mechanical transduction mechanisms associated with the development of glaucoma and may provide ideas into novel treatments for preventing the progression of glaucoma by targeting mechanotransduction.
Collapse
Affiliation(s)
- Lingling Jing
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Kexin Liu
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Feng Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Ying Su
- Eye Hospital, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
6
|
Cheng YW, Anzell AR, Morosky SA, Schwartze TA, Hinck CS, Hinck AP, Roman BL, Davidson LA. Shear Stress and Sub-Femtomolar Levels of Ligand Synergize to Activate ALK1 Signaling in Endothelial Cells. Cells 2024; 13:285. [PMID: 38334677 PMCID: PMC10854672 DOI: 10.3390/cells13030285] [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: 10/25/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Endothelial cells (ECs) respond to concurrent stimulation by biochemical factors and wall shear stress (SS) exerted by blood flow. Disruptions in flow-induced responses can result in remodeling issues and cardiovascular diseases, but the detailed mechanisms linking flow-mechanical cues and biochemical signaling remain unclear. Activin receptor-like kinase 1 (ALK1) integrates SS and ALK1-ligand cues in ECs; ALK1 mutations cause hereditary hemorrhagic telangiectasia (HHT), marked by arteriovenous malformation (AVM) development. However, the mechanistic underpinnings of ALK1 signaling modulation by fluid flow and the link to AVMs remain uncertain. We recorded EC responses under varying SS magnitudes and ALK1 ligand concentrations by assaying pSMAD1/5/9 nuclear localization using a custom multi-SS microfluidic device and a custom image analysis pipeline. We extended the previously reported synergy between SS and BMP9 to include BMP10 and BMP9/10. Moreover, we demonstrated that this synergy is effective even at extremely low SS magnitudes (0.4 dyn/cm2) and ALK1 ligand range (femtogram/mL). The synergistic response to ALK1 ligands and SS requires the kinase activity of ALK1. Moreover, ALK1's basal activity and response to minimal ligand levels depend on endocytosis, distinct from cell-cell junctions, cytoskeleton-mediated mechanosensing, or cholesterol-enriched microdomains. However, an in-depth analysis of ALK1 receptor trafficking's molecular mechanisms requires further investigation.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Anthony R. Anzell
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Stefanie A. Morosky
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tristin A. Schwartze
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cynthia S. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew P. Hinck
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lance A. Davidson
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
7
|
Chalkias A. Shear Stress and Endothelial Mechanotransduction in Trauma Patients with Hemorrhagic Shock: Hidden Coagulopathy Pathways and Novel Therapeutic Strategies. Int J Mol Sci 2023; 24:17522. [PMID: 38139351 PMCID: PMC10743945 DOI: 10.3390/ijms242417522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Massive trauma remains a leading cause of death and a global public health burden. Post-traumatic coagulopathy may be present even before the onset of resuscitation, and correlates with severity of trauma. Several mechanisms have been proposed to explain the development of abnormal coagulation processes, but the heterogeneity in injuries and patient profiles makes it difficult to define a dominant mechanism. Regardless of the pattern of death, a significant role in the pathophysiology and pathogenesis of coagulopathy may be attributed to the exposure of endothelial cells to abnormal physical forces and mechanical stimuli in their local environment. In these conditions, the cellular responses are translated into biochemical signals that induce/aggravate oxidative stress, inflammation, and coagulopathy. Microvascular shear stress-induced alterations could be treated or prevented by the development and use of innovative pharmacologic strategies that effectively target shear-mediated endothelial dysfunction, including shear-responsive drug delivery systems and novel antioxidants, and by targeting the venous side of the circulation to exploit the beneficial antithrombogenic profile of venous endothelial cells.
Collapse
Affiliation(s)
- Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5158, USA;
- Outcomes Research Consortium, Cleveland, OH 44195, USA
| |
Collapse
|
8
|
Jackson ML, Bond AR, George SJ. Mechanobiology of the endothelium in vascular health and disease: in vitro shear stress models. Cardiovasc Drugs Ther 2023; 37:997-1010. [PMID: 36190667 PMCID: PMC10516801 DOI: 10.1007/s10557-022-07385-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
Abstract
In recent years, there has been growing evidence that vascular pathologies arise in sites experiencing an altered haemodynamic environment. Fluid shear stress (FSS) is an important contributor to vascular homeostasis and regulates endothelial cell (EC) gene expression, morphology, and behaviour through specialised mechanosensitive signalling pathways. The presence of an altered FSS profile is a pathological characteristic of many vascular diseases, with the most established example being the preferential localisation of atherosclerotic plaque development. However, the precise haemodynamic contributions to other vascular pathologies including coronary artery vein graft failure remains poorly defined. To evaluate potential novel therapeutics for the treatment of vascular diseases via targeting EC behaviour, it is important to undertake in vitro experiments using appropriate culture conditions, particularly FSS. There are a wide range of in vitro models used to study the effect of FSS on the cultured endothelium, each with the ability to generate FSS flow profiles through which the investigator can control haemodynamic parameters including flow magnitude and directionality. An important consideration for selection of an appropriate model of FSS exposure is the FSS profile that the model can generate, in comparison to the physiological and pathophysiological haemodynamic environment of the vessel of interest. A resource bringing together the haemodynamic environment characteristic of atherosclerosis pathology and the flow profiles generated by in vitro methods of applying FSS would be beneficial to researchers when selecting the appropriate model for their research. Consequently, here we summarise the widely used methods of exposing cultured endothelium to FSS, the flow profile they generate and their advantages and limitations in investigating the pathological contribution of altered FSS to vascular disease and evaluating novel therapeutic targets for the treatment and prevention of vascular disease.
Collapse
Affiliation(s)
- Molly L. Jackson
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| | - Andrew Richard Bond
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| | - Sarah Jane George
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS2 8HW UK
| |
Collapse
|
9
|
Zhang F, Wu J, Shen Q, Chen Z, Qiao Z. Investigating the mechanism of Tongqiao Huoxue decotion in the treatment of allergic rhinitis based on network pharmacology and molecular docking: A review. Medicine (Baltimore) 2023; 102:e33190. [PMID: 36897696 PMCID: PMC9997813 DOI: 10.1097/md.0000000000033190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
Allergic rhinitis is prone to recurrence, and clinical treatments focus on control symptoms; however there is no radical cure. Our aim was to use network pharmacology and molecular docking to reveal the hub genes, biological functions, and signaling pathways of Tongqiao Huoxue decoction against allergic rhinitis. First, the chemical components and target genes of Tongqiao Huoxue decoction were obtained from the Traditional Chinese Medicine Systems Pharmacology database. Similarly, allergic rhinitis targets were screened using online Mendelian Inheritance In Man and GeneCards database. Then, all potential targets of Tongqiao Huoxue decoction in the treatment of allergic rhinitis were identified, the Venn diagram was portrayed using R software, and protein-protein interaction network was built using String. The hub genes were analyzed using enrichment analyses. Finally, molecular docking was used to verify the reliability of the key gene prediction. The core targets for Tongqiao Huoxue decoction to improve allergic rhinitis were AKT1, TP53, IL6, and so on. The enrichment analysis results showed that Tongqiao Huoxue decoction treatment in allergic rhinitis might be involved in the AGE-RAGE signaling pathway and fluid shear stress and atherosclerosis pathway. The molecular docking verification indicated that its ingredients bound well to the core targets of allergic rhinitis, and stigmasterol's docking ability with TNF (-12.73 kcal/mol) is particularly prominent. Based on these findings, it may be deduced that stigmasterol treated allergic rhinitis by acting on TNF targets. But, this conclusion needs to be confirmed by further in vitro and in vivo trials.
Collapse
Affiliation(s)
- Fang Zhang
- The First Clinical Medical College of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Jiani Wu
- Department of Otorhinolaryngology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qu Shen
- The First Clinical Medical College of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Zhiling Chen
- Department of Otorhinolaryngology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Zukang Qiao
- Department of Tuina, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
10
|
Soto Veliz D, Lin K, Sahlgren C. Organ-on-a-chip technologies for biomedical research and drug development: A focus on the vasculature. SMART MEDICINE 2023; 2:e20220030. [PMID: 37089706 PMCID: PMC7614466 DOI: 10.1002/smmd.20220030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/20/2023] [Indexed: 04/25/2023]
Abstract
Current biomedical models fail to replicate the complexity of human biology. Consequently, almost 90% of drug candidates fail during clinical trials after decades of research and billions of investments in drug development. Despite their physiological similarities, animal models often misrepresent human responses, and instead, trigger ethical and societal debates regarding their use. The overall aim across regulatory entities worldwide is to replace, reduce, and refine the use of animal experimentation, a concept known as the Three Rs principle. In response, researchers develop experimental alternatives to improve the biological relevance of in vitro models through interdisciplinary approaches. This article highlights the emerging organ-on-a-chip technologies, also known as microphysiological systems, with a focus on models of the vasculature. The cardiovascular system transports all necessary substances, including drugs, throughout the body while in charge of thermal regulation and communication between other organ systems. In addition, we discuss the benefits, limitations, and challenges in the widespread use of new biomedical models. Coupled with patient-derived induced pluripotent stem cells, organ-on-a-chip technologies are the future of drug discovery, development, and personalized medicine.
Collapse
Affiliation(s)
- Diosangeles Soto Veliz
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Kai‐Lan Lin
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Cecilia Sahlgren
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
11
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
12
|
Endothelial mechanosensing: A forgotten target to treat vascular remodeling in hypertension? Biochem Pharmacol 2022; 206:115290. [DOI: 10.1016/j.bcp.2022.115290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
|
13
|
Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022; 186:114319. [PMID: 35545136 DOI: 10.1016/j.addr.2022.114319] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on immune checkpoint inhibitors has evolved into a new pillar of cancer treatment in clinics, but dealing with treatment resistance (either primary or acquired) is a major challenge. The tumor microenvironment (TME) has a substantial impact on the pathological behaviors and treatment response of many cancers. The biophysical clues in TME have recently been considered as important characteristics of cancer. Furthermore, there is mounting evidence that biophysical cues in TME play important roles in each step of the cascade of cancer immunotherapy that synergistically contribute to immunotherapy resistance. In this review, we summarize five main biophysical cues in TME that affect resistance to immunotherapy: extracellular matrix (ECM) structure, ECM stiffness, tumor interstitial fluid pressure (IFP), solid stress, and vascular shear stress. First, the biophysical factors involved in anti-tumor immunity and therapeutic antibody delivery processes are reviewed. Then, the causes of these five biophysical cues and how they contribute to immunotherapy resistance are discussed. Finally, the latest treatment strategies that aim to improve immunotherapy efficacy by targeting these biophysical cues are shared. This review highlights the biophysical cues that lead to immunotherapy resistance, also supplements their importance in related technologies for studying TME biophysical cues in vitro and therapeutic strategies targeting biophysical cues to improve the effects of immunotherapy.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuanbo Jia
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yang Yu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
14
|
González A, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MY, Aimo A, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Delgado V, Emdin M, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund PS, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, Van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Bayés-Genís A. Cardiac remodelling - Part 1: From cells and tissues to circulating biomarkers. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:927-943. [PMID: 35334137 DOI: 10.1002/ejhf.2493] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiac remodelling refers to changes in left ventricular structure and function over time, with a progressive deterioration that may lead to heart failure (HF) development (adverse remodelling) or vice versa a recovery (reverse remodelling) in response to HF treatment. Adverse remodelling predicts a worse outcome, whilst reverse remodelling predicts a better prognosis. The geometry, systolic and diastolic function and electric activity of the left ventricle are affected, as well as the left atrium and on the long term even right heart chambers. At a cellular and molecular level, remodelling involves all components of cardiac tissue: cardiomyocytes, fibroblasts, endothelial cells and leucocytes. The molecular, cellular and histological signatures of remodelling may differ according to the cause and severity of cardiac damage, and clearly to the global trend toward worsening or recovery. These processes cannot be routinely evaluated through endomyocardial biopsies, but may be reflected by circulating levels of several biomarkers. Different classes of biomarkers (e.g. proteins, non-coding RNAs, metabolites and/or epigenetic modifications) and several biomarkers of each class might inform on some aspects on HF development, progression and long-term outcomes, but most have failed to enter clinical practice. This may be due to the biological complexity of remodelling, so that no single biomarker could provide great insight on remodelling when assessed alone. Another possible reason is a still incomplete understanding of the role of biomarkers in the pathophysiology of cardiac remodelling. Such role will be investigated in the first part of this review paper on biomarkers of cardiac remodelling.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - A Mark Richards
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand
| | - Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) and Rebirth Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Inês Falcao-Pires
- Department od Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Departments of Cardiology and Cardiac Surgery, and Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Roger S Y Foo
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Mark Y Chan
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Chukwuemeka G Anene-Nzelu
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Montreal Heart Institute, Montreal, Canada
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- Universitätsklinikum des Saarlandes, Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Saarland University, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Rigshospitalet-Copenhagen University Hospital, Heart Centre, Department of Cardiology, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | - Pardeep S Jhund
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Portugal
| | | | | | - Julio Núñez
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie Van Linthout
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Kant S, Tran KV, Kvandova M, Caliz AD, Yoo HJ, Learnard H, Dolan AC, Craige SM, Hall JD, Jiménez JM, St. Hilaire C, Schulz E, Kröller-Schön S, Keaney JF. PGC1α Regulates the Endothelial Response to Fluid Shear Stress via Telomerase Reverse Transcriptase Control of Heme Oxygenase-1. Arterioscler Thromb Vasc Biol 2022; 42:19-34. [PMID: 34789002 PMCID: PMC8702461 DOI: 10.1161/atvbaha.121.317066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Fluid shear stress (FSS) is known to mediate multiple phenotypic changes in the endothelium. Laminar FSS (undisturbed flow) is known to promote endothelial alignment to flow, which is key to stabilizing the endothelium and rendering it resistant to atherosclerosis and thrombosis. The molecular pathways responsible for endothelial responses to FSS are only partially understood. In this study, we determine the role of PGC1α (peroxisome proliferator gamma coactivator-1α)-TERT (telomerase reverse transcriptase)-HMOX1 (heme oxygenase-1) during shear stress in vitro and in vivo. Approach and Results: Here, we have identified PGC1α as a flow-responsive gene required for endothelial flow alignment in vitro and in vivo. Compared with oscillatory FSS (disturbed flow) or static conditions, laminar FSS (undisturbed flow) showed increased PGC1α expression and its transcriptional coactivation. PGC1α was required for laminar FSS-induced expression of TERT in vitro and in vivo via its association with ERRα(estrogen-related receptor alpha) and KLF (Kruppel-like factor)-4 on the TERT promoter. We found that TERT inhibition attenuated endothelial flow alignment, elongation, and nuclear polarization in response to laminar FSS in vitro and in vivo. Among the flow-responsive genes sensitive to TERT status, HMOX1 was required for endothelial alignment to laminar FSS. CONCLUSIONS These data suggest an important role for a PGC1α-TERT-HMOX1 axis in the endothelial stabilization response to laminar FSS.
Collapse
Affiliation(s)
- Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Equal contribution
| | - Khanh-Van Tran
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
- Equal contribution
| | - Miroslava Kvandova
- Department of Cardiology, University Medical Center, Mainz, Germany
- Equal contribution
| | - Amada D. Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Hyung-Jin Yoo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Heather Learnard
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Ana C. Dolan
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Siobhan M. Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blackburg, VA 24061
| | - Joshua D. Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Juan M. Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Cynthia St. Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, and the Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261
| | - Eberhard Schulz
- Department of Cardiology, Allgemeines Krankenhaus, Celle, Germany
| | | | - John F. Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
16
|
Yi S, Yang Y. Melatonin attenuates low shear stress-induced pyroptosis and endothelial cell dysfunction via the RORα/miR-223/STAT-3 signalling pathway. Exp Ther Med 2021; 22:1392. [PMID: 34650640 PMCID: PMC8506941 DOI: 10.3892/etm.2021.10828] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
Endothelial cells sense changes in blood flow shear stress and affect the progression of atherosclerotic plaques. Pyroptosis is an inflammatory form of cell death and has been implicated in cardiovascular diseases. Melatonin and its nuclear receptor retinoid-related orphan receptor α (RORα) have protective effects on the development of atherosclerosis. To date, whether melatonin can prevent endothelial cell pyroptosis and dysfunction in pathological shear stress remains unclear. In the present study, human umbilical vein endothelial cells (ECs) were cultured under low shear stress conditions (5 dyne/cm2) for 24 h and treated with or without melatonin (2 µmol/l). The binding sites of the microRNA (miR)-223 promoter and RORα were predicted using the JASPAR website. Expression of pyroptosis-related proteins, including cleaved N-terminal gasdermin D, caspase-1, intercellular adhesion molecule 1 (ICAM-1) and nitric oxide (NO) were assessed. The results indicated that low shear stress increased pyroptosis and ICAM-1 expression, whereas it decreased NO levels. Melatonin alleviated pyroptosis and ICAM-1 expression and increased the production of NO in ECs. Further assessment revealed that low-level shear stress decreased RORα protein and mRNA expression, whereas melatonin would bind to RORα and thereby promoted miR-223 transcription in ECs. The present study also identified signal transducer and activator of transcription 3 (STAT-3) as a potential target gene of miR-223-3p. When transfected with miR-223 inhibitor, ECs up-regulated the expression of pyroptosis-related proteins and ICAM-1, and down-regulated NO levels. By contrast, silencing STAT-3 expression diminished the protective effect of miR-223. These results indicated that melatonin prevented ECs from undergoing pyroptosis and alleviated dysfunction via the RORα/miR-223/STAT-3 signalling pathway. This information could aid in the development of novel therapeutic approaches and provide new insights into atherosclerosis.
Collapse
Affiliation(s)
- Sui Yi
- The Intensive Care Unit Department, Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Yang Yang
- The Neurology Department, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
17
|
Dendrobium catenatum Lindl. Water Extracts Attenuate Atherosclerosis. Mediators Inflamm 2021; 2021:9951946. [PMID: 34475805 PMCID: PMC8407999 DOI: 10.1155/2021/9951946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/07/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Dendrobium catenatum Lindl. (DH) is a Chinese herbal medicine, which is often used to make tea to improve immunity in China. Rumor has it that DH has a protective effect against cardiovascular disease. However, it is not clear how DH can prevent cardiovascular disease, such as atherosclerosis (AS). Therefore, the purpose of this study is to study whether DH can prevent AS and the underlying mechanisms. Methods Zebrafish larvae were fed with high-cholesterol diet (HCD) to establish a zebrafish AS model. Then, we used DH water extracts (DHWE) to pretreat AS zebrafish. The plaque formation was detected by HE, EVG, and oil red O staining. Neutrophil and macrophage counts were calculated to evaluate the inflammation level. Reactive oxygen species (ROS) activity, malondialdehyde (MDA) content, and superoxide dismutase (SOD) activity in zebrafish were measured to reflect oxidative stress. The cholesterol accumulation and the levels of lipid, triglyceride (TG), and total cholesterol (TC) were measured to reflect lipid metabolism disorder. Then, parallel flow chamber was utilized to establish a low shear stress- (LSS-) induced endothelial cell (EC) dysfunction model. EA.hy926 cells were exposed to LSS (3 dyn/cm2) for 30 min and treated with DHWE. The levels of ROS, SOD, MDA, glutathione (GSH), and glutathiol (GSSG) in EA.hy926 cells were analysed to determine oxidative stress. The release of nitric oxide (NO), endothelin-1 (ET-1), and epoprostenol (PGI2) in EA.hy926 cells was measured to reflect EC dysfunction. The mRNA expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in EA.hy926 cells was detected to reflect EC dysfunction inflammation. Results The results showed that DHWE significantly reduced cholesterol accumulation and macrophage infiltration in early AS. Finally, DHWE significantly alleviate the lipid metabolism disorder, oxidative stress, and inflammation to reduce the plaque formation of AS zebrafish larval model. Meanwhile, we also found that DHWE significantly improved LSS-induced EC dysfunction and oxidative stress in vitro. Conclusion Our results indicate that DHWE could be used as a prevention method to prevent AS.
Collapse
|
18
|
Mahendra Y, He M, Rouf MA, Tjakra M, Fan L, Wang Y, Wang G. Progress and prospects of mechanotransducers in shear stress-sensitive signaling pathways in association with arteriovenous malformation. Clin Biomech (Bristol, Avon) 2021; 88:105417. [PMID: 34246943 DOI: 10.1016/j.clinbiomech.2021.105417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023]
Abstract
Arteriovenous malformations are congenital vascular lesions characterized by a direct and tangled connection between arteries and veins, which disrupts oxygen circulation and normal blood flow. Arteriovenous malformations often occur in the patient with hereditary hemorrhagic telangiectasia. The attempts to elucidate the causative factors and pathogenic mechanisms of arteriovenous malformations are now still in progress. Some studies reported that shear stress in blood flow is one of the factors involved in arteriovenous malformations manifestation. Through several mechanotransducers harboring the endothelial cells membrane, the signal from shear stress is transduced towards the responsible signaling pathways in endothelial cells to maintain cell homeostasis. Any disruption in this well-established communication will give rise to abnormal endothelial cells differentiation and specification, which will later promote arteriovenous malformations. In this review, we discuss the update of several mechanotransducers that have essential roles in shear stress-induced signaling pathways, such as activin receptor-like kinase 1, Endoglin, Notch, vascular endothelial growth factor receptor 2, Caveolin-1, Connexin37, and Connexin40. Any disruption of these signaling potentially causes arteriovenous malformations. We also present some recent insights into the fundamental analysis, which attempts to determine potential and alternative solutions to battle arteriovenous malformations, especially in a less invasive and risky way, such as gene treatments.
Collapse
Affiliation(s)
- Yoga Mahendra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Mei He
- Chongqing University Cancer Hospital, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing, China
| | - Muhammad Abdul Rouf
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Marco Tjakra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Longling Fan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
19
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
20
|
Mechanosensation and Mechanotransduction by Lymphatic Endothelial Cells Act as Important Regulators of Lymphatic Development and Function. Int J Mol Sci 2021; 22:ijms22083955. [PMID: 33921229 PMCID: PMC8070425 DOI: 10.3390/ijms22083955] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Our understanding of the function and development of the lymphatic system is expanding rapidly due to the identification of specific molecular markers and the availability of novel genetic approaches. In connection, it has been demonstrated that mechanical forces contribute to the endothelial cell fate commitment and play a critical role in influencing lymphatic endothelial cell shape and alignment by promoting sprouting, development, maturation of the lymphatic network, and coordinating lymphatic valve morphogenesis and the stabilization of lymphatic valves. However, the mechanosignaling and mechanotransduction pathways involved in these processes are poorly understood. Here, we provide an overview of the impact of mechanical forces on lymphatics and summarize the current understanding of the molecular mechanisms involved in the mechanosensation and mechanotransduction by lymphatic endothelial cells. We also discuss how these mechanosensitive pathways affect endothelial cell fate and regulate lymphatic development and function. A better understanding of these mechanisms may provide a deeper insight into the pathophysiology of various diseases associated with impaired lymphatic function, such as lymphedema and may eventually lead to the discovery of novel therapeutic targets for these conditions.
Collapse
|
21
|
Li AF, Tan L, Zhang S, Tao J, Wang Z, Wei D. Low shear stress-induced endothelial mesenchymal transformation via the down-regulation of TET2. Biochem Biophys Res Commun 2021; 545:20-26. [PMID: 33535102 DOI: 10.1016/j.bbrc.2021.01.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Atherosclerotic cardiovascular disease is the major cause of death worldwide. Low shear stress plays key roles on the initiation and progression of atherosclerosis (As). However, its underlying mechanism remains unclear. In this study, the effect of low shear stress on endothelial mesenchymal transformation (EndMT) and its underlying mechanism were explored. Results showed that in cultured human umbilical vein endothelial cells, low shear stress down-regulated the expression of TET2 and promoted EndMT. Loss of TET2 promoted EndMT with the Wnt/β-catenin signaling pathway. The enhancement in EndMT induced by low shear stress was attenuated by TET2 overexpression. In apoE-/- mice subjected to carotid artery local ligation, the EndMT and atherosclerotic lesions induced by low shear stress was attenuated by TET2 overexpression. Taken together, low shear stress promoted EndMT through the down-regulation of TET2, indicating that intervention with EndMT or the up-regulation of TET2 might be an alternative strategy for preventing As.
Collapse
Affiliation(s)
- AFang Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China; School of Nursing, Xiangnan University, Chenzhou, Hunan, 423000, China
| | - LiLan Tan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China
| | - ShuLei Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China
| | - Jun Tao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
22
|
Senevirathne SWMAI, Hasan J, Mathew A, Woodruff M, Yarlagadda PKDV. Bactericidal efficiency of micro- and nanostructured surfaces: a critical perspective. RSC Adv 2021; 11:1883-1900. [PMID: 35424086 PMCID: PMC8693530 DOI: 10.1039/d0ra08878a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/12/2020] [Indexed: 12/21/2022] Open
Abstract
Micro/nanostructured surfaces (MNSS) have shown the ability to inactivate bacterial cells by physical means. An enormous amount of research has been conducted in this area over the past decade. Here, we review the various surface factors that affect the bactericidal efficiency. For example, surface hydrophobicity of the substrate has been accepted to be influential on the bactericidal effect of the surface, but a review of the literature suggests that the influence of hydrophobicity differs with the bacterial species. Also, various bacterial viability quantification methods on MNSS are critically reviewed for their suitability for the purpose, and limitations of currently used protocols are discussed. Presently used static bacterial viability assays do not represent the conditions of which those surfaces could be applied. Such application conditions do have overlaying fluid flow, and bacterial behaviours are drastically different under flow conditions compared to under static conditions. Hence, it is proposed that the bactericidal effect should be assessed under relevant fluid flow conditions with factors such as shear stress and flowrate given due significance. This review will provide a range of opportunities for future research in design and engineering of micro/nanostructured surfaces with varying experimental conditions.
Collapse
Affiliation(s)
- S W M A I Senevirathne
- Science and Engineering Faculty, Queensland University of Technology (QUT) Brisbane Qld 4000 Australia
- Institute of Health and Biomedical Innovations 60 Musk Ave. Kelvin Grove Qld 4059 Australia
| | - J Hasan
- Science and Engineering Faculty, Queensland University of Technology (QUT) Brisbane Qld 4000 Australia
- Institute of Health and Biomedical Innovations 60 Musk Ave. Kelvin Grove Qld 4059 Australia
| | - A Mathew
- Science and Engineering Faculty, Queensland University of Technology (QUT) Brisbane Qld 4000 Australia
- Institute of Health and Biomedical Innovations 60 Musk Ave. Kelvin Grove Qld 4059 Australia
| | - M Woodruff
- Science and Engineering Faculty, Queensland University of Technology (QUT) Brisbane Qld 4000 Australia
- Institute of Health and Biomedical Innovations 60 Musk Ave. Kelvin Grove Qld 4059 Australia
| | - P K D V Yarlagadda
- Science and Engineering Faculty, Queensland University of Technology (QUT) Brisbane Qld 4000 Australia
- Institute of Health and Biomedical Innovations 60 Musk Ave. Kelvin Grove Qld 4059 Australia
| |
Collapse
|
23
|
Ricard N, Bailly S, Guignabert C, Simons M. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol 2021; 18:565-580. [PMID: 33627876 PMCID: PMC7903932 DOI: 10.1038/s41569-021-00517-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Endothelial cells are at the interface between circulating blood and tissues. This position confers on them a crucial role in controlling oxygen and nutrient exchange and cellular trafficking between blood and the perfused organs. The endothelium adopts a structure that is specific to the needs and function of each tissue and organ and is subject to tissue-specific signalling input. In adults, endothelial cells are quiescent, meaning that they are not proliferating. Quiescence was considered to be a state in which endothelial cells are not stimulated but are instead slumbering and awaiting activating signals. However, new evidence shows that quiescent endothelium is fully awake, that it constantly receives and initiates functionally important signalling inputs and that this state is actively regulated. Signalling pathways involved in the maintenance of functionally quiescent endothelia are starting to be identified and are a combination of endocrine, autocrine, paracrine and mechanical inputs. The paracrine pathways confer a microenvironment on the endothelial cells that is specific to the perfused organs and tissues. In this Review, we present the current knowledge of organ-specific signalling pathways involved in the maintenance of endothelial quiescence and the pathologies associated with their disruption. Linking organ-specific pathways and human vascular pathologies will pave the way towards the development of innovative preventive strategies and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Nicolas Ricard
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Sabine Bailly
- grid.457348.9Université Grenoble Alpes, INSERM, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France
| | - Christophe Guignabert
- grid.414221.0INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Michael Simons
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA ,grid.47100.320000000419368710Department of Cell Biology, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
24
|
Swain SM, Liddle RA. Piezo1 acts upstream of TRPV4 to induce pathological changes in endothelial cells due to shear stress. J Biol Chem 2020; 296:100171. [PMID: 33298523 PMCID: PMC7948745 DOI: 10.1074/jbc.ra120.015059] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022] Open
Abstract
The ion channels Piezo1 and TRPV4 have both, independently, been implicated in high venous pressure- and fluid shear stress-induced vascular hyperpermeability in endothelial cells. However, the mechanism by which Piezo1 and TRPV4 channels execute the same function is poorly understood. Here we demonstrate that Piezo1 regulates TRPV4 channel activation in endothelial cells and that Piezo1-mediated TRPV4 channel opening is a function of the strength and duration of fluid shear stress. We first confirmed that either fluid shear stress or the Piezo1 agonist, Yoda1, led to an elevation in intracellular calcium ([Ca2+]i) and that application of the Piezo1 antagonist, GsMTx4, completely blocked this change. We discovered that high and prolonged shear stress caused sustained [Ca2+]i elevation that was blocked by inhibition of TRPV4 channel opening. Moreover, Piezo1 stimulated TRPV4 opening through activation of phospholipase A2. TRPV4-dependent sustained [Ca2+]i elevation was responsible for fluid shear stress-mediated and Piezo1-mediated disruption of adherens junctions and actin remodeling. Blockade of TRPV4 channels with the selective TRPV4 blocker, HC067047, prevented the loss of endothelial cell integrity and actin disruption induced by Yoda1 or shear stress and prevented Piezo1-induced monocyte adhesion to endothelial cell monolayers. These findings demonstrate that Piezo1 activation by fluid shear stress initiates a calcium signal that causes TRPV4 opening, which in turn is responsible for the sustained phase calcium elevation that triggers pathological events in endothelial cells. Thus, deleterious effects of shear stress are initiated by Piezo1 but require TRPV4.
Collapse
Affiliation(s)
- Sandip M Swain
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Rodger A Liddle
- Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Veterans Affairs Health Care System, Durham, North Carolina, USA.
| |
Collapse
|
25
|
Lupu F, Kinasewitz G, Dormer K. The role of endothelial shear stress on haemodynamics, inflammation, coagulation and glycocalyx during sepsis. J Cell Mol Med 2020; 24:12258-12271. [PMID: 32951280 PMCID: PMC7687012 DOI: 10.1111/jcmm.15895] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a multifactorial syndrome primarily determined by the host response to an invading pathogen. It is common, with over 48 million cases worldwide in 2017, and often lethal. The sequence of events in sepsis begins with the damage of endothelium within the microvasculature, as a consequence of the inflammatory and coagulopathic responses to the pathogen that can progress to multiple organ failure and death. Most therapeutic interventions target the inflammation and coagulation pathways that act as an auto-amplified vicious cycle, which, if unchecked can be fatal. Normal blood flow and shear stress acting on a healthy endothelium and intact glycocalyx have anti-inflammatory, anticoagulant and self-repairing effects. During early stages of sepsis, the vascular endothelium and its glycocalyx become dysfunctional, yet they are essential components of resuscitation and recovery from sepsis. The effects of shear forces on sepsis-induced endothelial dysfunction, including inflammation, coagulation, complement activation and microcirculatory breakdown are reviewed. It is suggested that early therapeutic strategies should prioritize on the restoration of shear forces and endothelial function and on the preservation of the endothelial-glycocalyx barrier.
Collapse
Affiliation(s)
- Florea Lupu
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Gary Kinasewitz
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | | |
Collapse
|
26
|
Protective Activities of Dendrobium huoshanense C. Z. Tang et S. J. Cheng Polysaccharide against High-Cholesterol Diet-Induced Atherosclerosis in Zebrafish. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8365056. [PMID: 32724495 PMCID: PMC7366212 DOI: 10.1155/2020/8365056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/20/2020] [Indexed: 11/17/2022]
Abstract
Cardiovascular disease is the highest cause of death, and atherosclerosis (AS) is the primary pathogenesis of many cardiovascular diseases. In this study, we aim to investigate the possible pharmaceutical effects of Dendrobium huoshanense C. Z. Tang et S. J. Cheng polysaccharide (DHP) in AS. We fed zebrafish with high-cholesterol diet (HCD) to establish a zebrafish AS model and treated with DHP and observed plaque formation and neutrophil counts under a fluorescence microscope. Next, a parallel flow chamber was utilized to establish low shear stress- (LSS-) induced endothelial cell (EC) dysfunction model. We observed that DHP significantly improved HCD-induced lipid deposition, oxidative stress, and inflammatory response, mainly showing that DHP significantly increased superoxide dismutase (SOD) activity, decreased plaque formation, and decreased neutrophil recruitment and the levels of total cholesterol (TC), triglyceride (TG), malondialdehyde (MDA), and reactive oxygen species (ROS). Furthermore, DHP significantly improved LSS-induced oxidative stress and EC dysfunction. Our results indicated that DHP can exert treatment effects on AS, which may attribute to its hypolipidemic, antioxidant, anti-inflammatory activities and improving LSS-induced EC dysfunction. DHP has promising potential for further development as a functional natural medicine source targeted at AS prevention.
Collapse
|
27
|
Tian GE, Zhou JT, Liu XJ, Huang YC. Mechanoresponse of stem cells for vascular repair. World J Stem Cells 2019; 11:1104-1114. [PMID: 31875871 PMCID: PMC6904862 DOI: 10.4252/wjsc.v11.i12.1104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/25/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Stem cells have shown great potential in vascular repair. Numerous evidence indicates that mechanical forces such as shear stress and cyclic strain can regulate the adhesion, proliferation, migration, and differentiation of stem cells via serious signaling pathways. The enrichment and differentiation of stem cells play an important role in the angiogenesis and maintenance of vascular homeostasis. In normal tissues, blood flow directly affects the microenvironment of vascular endothelial cells (ECs); in pathological status, the abnormal interactions between blood flow and vessels contribute to the injury of vessels. Next, the altered mechanical forces are transduced into cells by mechanosensors to trigger the reformation of vessels. This process occurs when signaling pathways related to EC differentiation are initiated. Hence, a deep understanding of the responses of stem cells to mechanical stresses and the underlying mechanisms involved in this process is essential for clinical translation. In this the review, we provide an overview of the role of stem cells in vascular repair, outline the performance of stem cells under the mechanical stress stimulation, and describe the related signaling pathways.
Collapse
Affiliation(s)
- Ge-Er Tian
- Regenerative Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jun-Teng Zhou
- Department of Cardiology of West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xiao-Jing Liu
- Regenerative Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China.
| |
Collapse
|
28
|
Laschke MW, Kontaxi E, Scheuer C, Heß A, Karschnia P, Menger MD. Insulin-like growth factor 1 stimulates the angiogenic activity of adipose tissue-derived microvascular fragments. J Tissue Eng 2019; 10:2041731419879837. [PMID: 31632630 PMCID: PMC6767710 DOI: 10.1177/2041731419879837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis in adipose tissue is promoted by insulin-like growth factor 1 signaling. We analyzed whether this regulatory mechanism also improves the angiogenic activity of adipose tissue-derived microvascular fragments. Murine adipose tissue-derived microvascular fragments were cultivated for 24 h in the University of Wisconsin (UW) solution supplemented with vehicle, insulin-like growth factor 1, or a combination of insulin-like growth factor 1 and insulin-like growth factor-binding protein 4. Subsequently, we assessed their cellular composition, viability, proliferation, and growth factor expression. Moreover, cultivated adipose tissue-derived microvascular fragments were seeded onto collagen-glycosaminoglycan scaffolds, which were implanted into dorsal skinfold chambers to study their vascularization and incorporation. Insulin-like growth factor 1 increased the viability and growth factor expression of adipose tissue-derived microvascular fragments without affecting their cellular composition and proliferation. Accordingly, scaffolds containing insulin-like growth factor 1-stimulated adipose tissue-derived microvascular fragments exhibited an enhanced in vivo vascularization and incorporation. These positive insulin-like growth factor 1 effects were reversed by additional exposure of adipose tissue-derived microvascular fragments to insulin-like growth factor-binding protein 4. Our findings indicate that insulin-like growth factor 1 stimulation of adipose tissue-derived microvascular fragments is suitable to improve their vascularization capacity.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Elena Kontaxi
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Alexander Heß
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Philipp Karschnia
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|