1
|
Mensah EA, Daneshtalab N, Tabrizchi R. Effects of vasoactive substances on biomechanics of small resistance arteries of male and female Dahl salt-sensitive rats. Pharmacol Res Perspect 2024; 12:e1180. [PMID: 38421097 PMCID: PMC10902908 DOI: 10.1002/prp2.1180] [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: 11/14/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
Changes in vascular biomechanics leading to increase in arterial stiffness play a pivotal role in circulatory dysfunction. Our objectives were to examine sex-specific pharmacological changes related to the biomechanics and any structural modifications in small resistance arteries of Dahl salt-sensitive male and female rats. The composite Young modulus (CYM) was determined using pressure myograph recordings, and immunohistochemistry was used for the evaluation of any structural changes in the third-order mesenteric arteries (n = 6). Animals on high-salt diet developed hypertension with significant elevation in central and peripheral blood pressures and pulse wave velocity compared to those on regular diet. There were no significant differences observed in the CYM between any of the groups (i.e., males and females) in vehicle-treated time-control studies. The presence of verapamil (0.3 μM) significantly reduced CYM in hypertensive males without changes within females compared to vehicle. This effect was abolished by phenylephrine (0.3 μM). BaCl2 (100 μM), ouabain (100 μM), and L-NAME (0.3 μM) combined significantly increased CYM in vessels from in normotensive males and females but not in hypertensive males compared to vehicle. The increase in CYM was abolished in the presence of phenylephrine. Sodium nitroprusside (0.3 μM), in the presence of phenylephrine, significantly reduced CYM in male normotensive versus hypertensive, with no differences within females. Significant differences were observed in immunohistochemical assessment of biomechanical markers of arterial stiffness between males and females. Our findings suggest sex possibly due to pressure differences to be responsible for adaptive changes in biomechanics, and varied pharmacological responses in hypertensive state.
Collapse
Affiliation(s)
- Eric A. Mensah
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Noriko Daneshtalab
- School of PharmacyMemorial University NewfoundlandSt. John'sNewfoundlandCanada
| | - Reza Tabrizchi
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| |
Collapse
|
2
|
Bréchot N, Rutault A, Marangon I, Germain S. Blood endothelium transition and phenotypic plasticity: A key regulator of integrity/permeability in response to ischemia. Semin Cell Dev Biol 2024; 155:16-22. [PMID: 37479554 DOI: 10.1016/j.semcdb.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
In the human body, the 1013 blood endothelial cells (ECs) which cover a surface of 500-700 m2 (Mai et al., 2013) are key players of tissue homeostasis, remodeling and regeneration. Blood vessel ECs play a major role in the regulation of metabolic and gaz exchanges, cell trafficking, blood coagulation, vascular tone, blood flow and fluid extravasation (also referred to as blood vascular permeability). ECs are heterogeneous in various capillary beds and have the exquisite capacity to cope with environmental changes by regulating their gene expression. Ischemia has major detrimental effects on the endothelium and ischemia-induced regulation of vascular integrity is of paramount importance for human health, as small amounts of fluid accumulation in the interstitium may be responsible for major effects on organ functions and patients outcome. In this review, we will here focus on the stimuli and the molecular mechanisms that control blood endothelium maintenance and phenotypic plasticity/transition involved in controlling blood capillary leakage that might open new avenues for therapeutic applications.
Collapse
Affiliation(s)
- Nicolas Bréchot
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France; Intensive Care Medicine Department, Université de Paris Cité, Hôpital européen Georges-Pompidou, AP-HP, AP-HP.CUP, 75015 Paris, France.
| | - Alexandre Rutault
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Iris Marangon
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France.
| |
Collapse
|
3
|
Mutgan AC, Jandl K, Radic N, Valzano F, Kolb D, Hoffmann J, Foris V, Wilhelm J, Boehm PM, Hoetzenecker K, Olschewski A, Olschewski H, Heinemann A, Wygrecka M, Marsh LM, Kwapiszewska G. Pentastatin, a matrikine of the collagen IVα5, is a novel endogenous mediator of pulmonary endothelial dysfunction. Am J Physiol Cell Physiol 2023; 325:C1294-C1312. [PMID: 37694286 PMCID: PMC11550886 DOI: 10.1152/ajpcell.00391.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Deposition of basement membrane components, such as collagen IVα5, is associated with altered endothelial cell function in pulmonary hypertension. Collagen IVα5 harbors a functionally active fragment within its C-terminal noncollageneous (NC1) domain, called pentastatin, whose role in pulmonary endothelial cell behavior remains unknown. Here, we demonstrate that pentastatin serves as a mediator of pulmonary endothelial cell dysfunction, contributing to pulmonary hypertension. In vitro, treatment with pentastatin induced transcription of immediate early genes and proinflammatory cytokines and led to a functional loss of endothelial barrier integrity in pulmonary arterial endothelial cells. Mechanistically, pentastatin leads to β1-integrin subunit clustering and Rho/ROCK activation. Blockage of the β1-integrin subunit or the Rho/ROCK pathway partially attenuated the pentastatin-induced endothelial barrier disruption. Although pentastatin reduced the viability of endothelial cells, smooth muscle cell proliferation was induced. These effects on the pulmonary vascular cells were recapitulated ex vivo in the isolated-perfused lung model, where treatment with pentastatin-induced swelling of the endothelium accompanied by occasional endothelial cell apoptosis. This was reflected by increased vascular permeability and elevated pulmonary arterial pressure induced by pentastatin. This study identifies pentastatin as a mediator of endothelial cell dysfunction, which thus might contribute to the pathogenesis of pulmonary vascular disorders such as pulmonary hypertension.NEW & NOTEWORTHY This study is the first to show that pentastatin, the matrikine of the basement membrane (BM) collagen IVα5 polypeptide, triggers rapid pulmonary arterial endothelial cell barrier disruption, activation, and apoptosis in vitro and ex vivo. Mechanistically, pentastatin partially acts through binding to the β1-integrin subunit and the Rho/ROCK pathway. These findings are the first to link pentastatin to pulmonary endothelial dysfunction and, thus, suggest a major role for BM-matrikines in pulmonary vascular diseases such as pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Nemanja Radic
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Francesco Valzano
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Jochen Wilhelm
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| | - Panja M Boehm
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Malgorzata Wygrecka
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
- Center for Infection and Genomics of the Lung, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Leigh M Marsh
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
4
|
Nhàn NTT, Yamada T, Yamada KH. Peptide-Based Agents for Cancer Treatment: Current Applications and Future Directions. Int J Mol Sci 2023; 24:12931. [PMID: 37629112 PMCID: PMC10454368 DOI: 10.3390/ijms241612931] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Peptide-based strategies have received an enormous amount of attention because of their specificity and applicability. Their specificity and tumor-targeting ability are applied to diagnosis and treatment for cancer patients. In this review, we will summarize recent advancements and future perspectives on peptide-based strategies for cancer treatment. The literature search was conducted to identify relevant articles for peptide-based strategies for cancer treatment. It was performed using PubMed for articles in English until June 2023. Information on clinical trials was also obtained from ClinicalTrial.gov. Given that peptide-based strategies have several advantages such as targeted delivery to the diseased area, personalized designs, relatively small sizes, and simple production process, bioactive peptides having anti-cancer activities (anti-cancer peptides or ACPs) have been tested in pre-clinical settings and clinical trials. The capability of peptides for tumor targeting is essentially useful for peptide-drug conjugates (PDCs), diagnosis, and image-guided surgery. Immunomodulation with peptide vaccines has been extensively tested in clinical trials. Despite such advantages, FDA-approved peptide agents for solid cancer are still limited. This review will provide a detailed overview of current approaches, design strategies, routes of administration, and new technological advancements. We will highlight the success and limitations of peptide-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL 60607, USA
| | - Kaori H. Yamada
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology & Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Li C, Xu B, Song H, Xu Y, Shi LZ, Chen XQ, Song ZC. Calcium dobesilate prevents PLD-induced hand-foot syndrome by alleviating capillary endothelial tight junction injury via the HA/CD44 pathway. Am J Cancer Res 2023; 13:3234-3245. [PMID: 37559988 PMCID: PMC10408487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
Pegylated liposomal doxorubicin (PLD) has excellent therapeutic efficacy in the treatment of cancers, but can cause serious adverse reactions such as hand-foot syndrome (HFS). Our previous research suggests that both PLD-induced HFS may be associated with injury to tight junctions (TJs) in the skin and that calcium dobesilate (CaD) can alleviate HFS. However, the underlying molecular mechanism is not well understood. Here, we created an in vitro PLD-treated model using Human Microvascular Endothelial Cell line-1 (HMEC-1) and an in vivo HFS rat model to investigate the underlying pathways. Treatment with PLD increased the expression of HYAL-1, CD44, and hyaluronic acid (HA) concentration, while reducing ZO-1 and Claudin-5 expression. Moreover, PLD treatment induced the degradation of higher molecular weight HA to its lower molecular weight counterpart, elevating the permeability of both HEMC-1 cell membranes and rat paw skin capillaries. AD-01 (CD44 inhibitor) inhibited the effect of PLD on the expression of ZO-1 and Claudin-5. Furthermore, CaD treatment suppressed the expression of HYAL-1 and CD44, mitigated HA degradation, and enhanced the expression of ZO-1 and Claudin-5. This resulted in decreased permeability in HEMC-1 cells and rat skin capillaries. In summary, our data suggest that PLD may promote the destruction of TJs via the HA/CD44 pathway, thereby leading to HFS through increased skin permeability and exacerbated doxorubicin extravasation. Moreover, CaD can inhibit this pathway, offering a potential therapeutic avenue to alleviate HFS.
Collapse
Affiliation(s)
- Chao Li
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Bin Xu
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Heng Song
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Yu Xu
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Ling-Zi Shi
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Xiao-Qing Chen
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
| | - Zhen-Chuan Song
- Department of Breast Center, The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050000, Hebei, P. R. China
- Key Laboratory for Breast Cancer Molecular Medicine of Hebei ProvinceShijiazhuang 050000, Hebei, P. R. China
| |
Collapse
|
6
|
Chen W, Yu H, Sun C, Dong M, Zhao N, Wang Y, Yu K, Zhang J, Xu N, Liu W. γ-Bungarotoxin impairs the vascular endothelial barrier function by inhibiting integrin α5. Toxicol Lett 2023; 383:177-191. [PMID: 37392970 DOI: 10.1016/j.toxlet.2023.06.009] [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: 03/04/2023] [Revised: 06/06/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
γ-bungarotoxin (γ-BGT) is an RGD motif-containing protein, derived from the venom of Bungarus multicinctus, leading to acute death in mice. These RGD motif-containing proteins from snake venom belonging to the disintegrin family can interfere with vascular endothelial homeostasis by directly binding cell surface integrins. Targeting integrins that generate vascular endothelial dysfunction may contribute to γ-BGT poisoning, however, the underlying mechanisms have not been investigated in detail. In this study, the results showed that γ-BGT played a role in -promoting the permeability of the vascular endothelial barrier. Depending on its selective binding to integrin α5 in vascular endothelium (VE), γ-BGT initiated downstream events, including focal adhesion kinase dephosphorylation and cytoskeleton remodeling, resulting in the intercellular junction interruption. Those alternations facilitated paracellular permeability of VE and barrier dysfunction. Proteomics profiling identified that as a downstream effector of the integrin α5 / FAK signaling pathway cyclin D1 partially mediated the cellular structural changes and barrier dysfunction. Furthermore, VE-released plasminogen activator urokinase and platelet-derived growth factor D could serve as potential diagnostic biomarkers for γ-BGT-induced vascular endothelial dysfunction. Our results indicate the mechanisms through which γ-BGT as a novel disintegrin directly interacts with the VE, with consequences for barrier dysfunction.
Collapse
Affiliation(s)
- Wei Chen
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, Jilin, PR China
| | - Haotian Yu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Chengbiao Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Mingxin Dong
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Na Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Yan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Kaikai Yu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Jianxu Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China
| | - Na Xu
- Jilin Medical University, Jilin 132013, Jilin, PR China.
| | - Wensen Liu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, Jilin, PR China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, Jilin, PR China.
| |
Collapse
|
7
|
Kuwar R, Wen X, Zhang N, Sun D. Integrin binding peptides facilitate growth and interconnected vascular-like network formation of rat primary cortical vascular endothelial cells in vitro. Neural Regen Res 2023; 18:1052-1056. [PMID: 36254992 PMCID: PMC9827785 DOI: 10.4103/1673-5374.355760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022] Open
Abstract
Neovascularization and angiogenesis in the brain are important physiological processes for normal brain development and repair/regeneration following insults. Integrins are cell surface adhesion receptors mediating important function of cells such as survival, growth and development during tissue organization, differentiation and organogenesis. In this study, we used an integrin-binding array platform to identify the important types of integrins and their binding peptides that facilitate adhesion, growth, development, and vascular-like network formation of rat primary brain microvascular endothelial cells. Brain microvascular endothelial cells were isolated from rat brain on post-natal day 7. Cells were cultured in a custom-designed integrin array system containing short synthetic peptides binding to 16 types of integrins commonly expressed on cells in vertebrates. After 7 days of culture, the brain microvascular endothelial cells were processed for immunostaining with markers for endothelial cells including von Willibrand factor and platelet endothelial cell adhesion molecule. 5-Bromo-2'-dexoyuridine was added to the culture at 48 hours prior to fixation to assess cell proliferation. Among 16 integrins tested, we found that α5β1, αvβ5 and αvβ8 greatly promoted proliferation of endothelial cells in culture. To investigate the effect of integrin-binding peptides in promoting neovascularization and angiogenesis, the binding peptides to the above three types of integrins were immobilized to our custom-designed hydrogel in three-dimensional (3D) culture of brain microvascular endothelial cells with the addition of vascular endothelial growth factor. Following a 7-day 3D culture, the culture was fixed and processed for double labeling of phalloidin with von Willibrand factor or platelet endothelial cell adhesion molecule and assessed under confocal microscopy. In the 3D culture in hydrogels conjugated with the integrin-binding peptide, brain microvascular endothelial cells formed interconnected vascular-like network with clearly discernable lumens, which is reminiscent of brain microvascular network in vivo. With the novel integrin-binding array system, we identified the specific types of integrins on brain microvascular endothelial cells that mediate cell adhesion and growth followed by functionalizing a 3D hydrogel culture system using the binding peptides that specifically bind to the identified integrins, leading to robust growth and lumenized microvascular-like network formation of brain microvascular endothelial cells in 3D culture. This technology can be used for in vitro and in vivo vascularization of transplants or brain lesions to promote brain tissue regeneration following neurological insults.
Collapse
Affiliation(s)
- Ram Kuwar
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Ning Zhang
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Dong Sun
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
8
|
Aleksandrowicz R, Strączkowski M. Link between insulin resistance and skeletal muscle extracellular matrix remodeling. Endocr Connect 2023; 12:e230023. [PMID: 36917038 PMCID: PMC10160556 DOI: 10.1530/ec-23-0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/16/2023]
Abstract
Skeletal muscle is the main metabolic tissue responsible for glucose homeostasis in the body. It is surrounded by the extracellular matrix (ECM) consisting of three layers: epimysium, perimysium, and endomysium. ECM plays an important role in the muscle, as it provides integrity and scaffolding cells. The observed disturbances in this structure are related to the abnormal remodeling of the ECM (through an increase in the concentration of its components). ECM rearrangement may impair insulin action by increasing the physical barrier to insulin transport and reducing insulin transport into muscle cells as well as by directly inhibiting insulin action through integrin signaling. Thus, improper ECM remodeling may contribute to the development of insulin resistance (IR) and related comorbidities. In turn, IR-associated conditions may further aggravate disturbances of ECM in skeletal muscle. This review describes the major components of the ECM that are necessary for its proper function. Particular attention was also paid to receptors (integrins) involved in the signaling of metabolic pathways. Finally, changes in ECM components in the context of clinical and animal studies are discussed. This article will help the reader to systematize knowledge related to the ECM and to better understand the relationship between ECM remodeling and IR, and its role in the pathogenesis of T2DM. The information in this article presents the concept of the role of ECM and its remodeling in the pathogenesis of IR, which may contribute to developing new therapeutic solutions.
Collapse
Affiliation(s)
- Róża Aleksandrowicz
- Department of Prophylaxis of Metabolic Diseases, Bialystok, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Bialystok, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
9
|
Yang L, Wang M, Hu X, Yuan L, Chen S, Peng S, Yang P, Yang Z, Bao G, He X. EccDNA-oriented ITGB7 expression in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1344. [PMID: 36660685 PMCID: PMC9843317 DOI: 10.21037/atm-22-5716] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
Background Extrachromosomal circular DNA (eccDNA) is omnipresent in cancers and related to the progression of tumors and oncogene amplification. However, its function in breast cancer (BC) is unclear. Methods After constructing the DNA library, CLeavage Effects by Circularization for In vitro Reporting of sequencing was performed for eccDNA detection using 1 BC tissue sample. Fastqc was used to evaluate the quality of the original data. Burrows-Wheeler-Alignment Tool was used to compare the original data to the reference genome. A Circle-MAP was subsequently performed to detect eccDNA, and Bedtools was used to annotate the eccDNA genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were conducted by ClusterProfiler. The Genotype-Tissue Expression and the Cancer Genome Atlas databases were used to collect the ribonucleic acid-sequencing data of the BC and normal samples. A Gene Expression Profiling Interactive Analysis, the University of Alabama at Birmingham CANcer data analysis Portal, and Kaplan-Meier survival curves were used to analyze the Cancer Genome Atlas data. Results A total of 200 eccDNA genes, including IGTB7, were obtained. About the biological processes (BPs), these 200 genes were mainly enriched in actin cytoskeleton reorganization and axon guidance. Concerning the molecular functions (MFs), these 200 genes were mainly enriched in sodium ion transmembrane transporter activity and metal ion transmembrane transporter activity. As for cellular components (CCs), these 200 genes were mainly enriched in the transcription regulator complex and focal adhesion. ITGB7 was significantly enriched in cell-matrix adhesion and localization within the membrane in the BPs, integrin binding in the MFs, and cell-substrate junction and focal adhesion in the CCs. The 200 eccDNA genes were mainly enriched in the PI3K-Akt signaling pathway and focal adhesion. Notably, ITGB7 was enriched in focal adhesion, ECM-receptor interaction, the PI3K-Akt signaling pathway, and human papillomavirus infection. Besides, ITGB7 was significantly upregulated in BC patients and was associated with the menopause status of the BC patients. Conclusions ITGB7 might serve as a prognostic marker for BC patients. ITGB7 has important implications for the individualized clinical treatment of BC patients.
Collapse
Affiliation(s)
- Lin Yang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Meixue Wang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xi'e Hu
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Lijuan Yuan
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Songhao Chen
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Shujia Peng
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Ping Yang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Zhenyu Yang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Guoqiang Bao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xianli He
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| |
Collapse
|
10
|
Exarchos V, Zacharova E, Neuber S, Giampietro C, Motta SE, Hinkov H, Emmert MY, Nazari-Shafti TZ. The path to a hemocompatible cardiovascular implant: Advances and challenges of current endothelialization strategies. Front Cardiovasc Med 2022; 9:971028. [PMID: 36186971 PMCID: PMC9515323 DOI: 10.3389/fcvm.2022.971028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular (CV) implants are still associated with thrombogenicity due to insufficient hemocompatibility. Endothelialization of their luminal surface is a promising strategy to increase their hemocompatibility. In this review, we provide a collection of research studies and review articles aiming to summarize the recent efforts on surface modifications of CV implants, including stents, grafts, valves, and ventricular assist devises. We focus in particular on the implementation of micrometer or nanoscale surface modifications, physical characteristics of known biomaterials (such as wetness and stiffness), and surface morphological features (such as gratings, fibers, pores, and pits). We also review how biomechanical signals originating from the endothelial cell for surface interaction can be directed by topography engineering approaches toward the survival of the endothelium and its long-term adaptation. Finally, we summarize the regulatory and economic challenges that may prevent clinical implementation of endothelialized CV implants.
Collapse
Affiliation(s)
- Vasileios Exarchos
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Ema Zacharova
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Department of Life Sciences, IMC University of Applied Sciences Krems, Krems an der Donau, Austria
| | - Sebastian Neuber
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Costanza Giampietro
- Experimental Continuum Mechanics, Empa Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, Switzerland
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zürich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Hristian Hinkov
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian Y. Emmert
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Clinic for Cardiovascular Surgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Timo Z. Nazari-Shafti
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin, Germany
- *Correspondence: Timo Z. Nazari-Shafti,
| |
Collapse
|
11
|
van Schaik PEM, Zuhorn IS, Baron W. Targeting Fibronectin to Overcome Remyelination Failure in Multiple Sclerosis: The Need for Brain- and Lesion-Targeted Drug Delivery. Int J Mol Sci 2022; 23:8418. [PMID: 35955549 PMCID: PMC9368816 DOI: 10.3390/ijms23158418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease with unknown etiology that can be characterized by the presence of demyelinated lesions. Prevailing treatment protocols in MS rely on the modulation of the inflammatory process but do not impact disease progression. Remyelination is an essential factor for both axonal survival and functional neurological recovery but is often insufficient. The extracellular matrix protein fibronectin contributes to the inhibitory environment created in MS lesions and likely plays a causative role in remyelination failure. The presence of the blood-brain barrier (BBB) hinders the delivery of remyelination therapeutics to lesions. Therefore, therapeutic interventions to normalize the pathogenic MS lesion environment need to be able to cross the BBB. In this review, we outline the multifaceted roles of fibronectin in MS pathogenesis and discuss promising therapeutic targets and agents to overcome fibronectin-mediated inhibition of remyelination. In addition, to pave the way for clinical use, we reflect on opportunities to deliver MS therapeutics to lesions through the utilization of nanomedicine and discuss strategies to deliver fibronectin-directed therapeutics across the BBB. The use of well-designed nanocarriers with appropriate surface functionalization to cross the BBB and target the lesion sites is recommended.
Collapse
Affiliation(s)
- Pauline E. M. van Schaik
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| |
Collapse
|
12
|
Edgar LT, Park H, Crawshaw JR, Osborne JM, Eichmann A, Bernabeu MO. Traffic Patterns of the Migrating Endothelium: How Force Transmission Regulates Vascular Malformation and Functional Shunting During Angiogenic Remodelling. Front Cell Dev Biol 2022; 10:840066. [PMID: 35663401 PMCID: PMC9160721 DOI: 10.3389/fcell.2022.840066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis occurs in distinct phases: initial spouting is followed by remodelling in which endothelial cells (ECs) composing blood vessels rearrange by migrating against the direction of flow. Abnormal remodelling can result in vascular malformation. Such is the case in mutation of the Alk1 receptor within the mouse retina which disrupts flow-migration coupling, creating mixed populations of ECs polarised with/against flow which aggregate into arteriovenous malformations (AVMs). The lack of live imaging options in vivo means that the collective EC dynamics that drive AVM and the consequences of mixed populations of polarity remain a mystery. Therefore, our goal is to present a novel agent-based model to provide theoretical insight into EC force transmission and collective dynamics during angiogenic remodelling. Force transmission between neighbouring agents consists of extrusive forces which maintain spacing and cohesive forces which maintain the collective. We performed migration simulations within uniformly polarised populations (against flow) and mixed polarity (with/against flow). Within uniformly polarised populations, extrusive forces stabilised the plexus by facilitating EC intercalation which ensures that cells remained evenly distributed. Excess cohesion disrupts intercalation, resulting in aggregations of cells and functional shunting. Excess cohesion between ECs prevents them from resolving diameter balances within the plexus, leading to prolonged flow reversals which exert a critical behaviour change within the system as they switch the direction of cell migration and traffic patterns at bifurcations. Introducing mixtures of cell polarity dramatically changed the role of extrusive forces within the system. At low extrusion, opposing ECs were able to move past each other; however, at high extrusion the pushing between cells resulted in migration speeds close to zero, forming traffic jams and disrupting migration. In our study, we produced vascular malformations and functional shunting with either excess cohesion between ECs or mixtures of cell polarity. At the centre of both these mechanisms are cell-cell adherens junctions, which are involved in flow sensing/polarity and must remodelling dynamically to allow rearrangements of cells during vascular patterning. Thus, our findings implicate junctional dysfunction as a new target in the treatment and prevention of vascular disease and AVMs.
Collapse
Affiliation(s)
- Lowell T. Edgar
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lowell T. Edgar, ; Miguel O. Bernabeu,
| | - Hyojin Park
- Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jessica R. Crawshaw
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia
| | - James M. Osborne
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia
| | - Anne Eichmann
- Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Yale University School of Medicine, Department of Cellular and Molecular Physiology, New Haven, CT, United States
- Université de Paris, PARCC, INSERM, Paris, France
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lowell T. Edgar, ; Miguel O. Bernabeu,
| |
Collapse
|
13
|
Nader D, Kerrigan SW. Molecular Cross-Talk between Integrins and Cadherins Leads to a Loss of Vascular Barrier Integrity during SARS-CoV-2 Infection. Viruses 2022; 14:891. [PMID: 35632633 PMCID: PMC9143673 DOI: 10.3390/v14050891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
The vascular barrier is heavily injured following SARS-CoV-2 infection and contributes enormously to life-threatening complications in COVID-19. This endothelial dysfunction is associated with the phlogistic phenomenon of cytokine storms, thrombotic complications, abnormal coagulation, hypoxemia, and multiple organ failure. The mechanisms surrounding COVID-19 associated endotheliitis have been widely attributed to ACE2-mediated pathways. However, integrins are emerging as possible receptor candidates for SARS-CoV-2, and their complex intracellular signaling events are essential for maintaining endothelial homeostasis. Here, we showed that the spike protein of SARS-CoV-2 depends on its RGD motif to drive barrier dysregulation by hijacking integrin αVβ3, expressed on human endothelial cells. This triggers the redistribution and internalization of major junction protein VE-Cadherin which leads to the barrier disruption phenotype. Both extracellular and intracellular inhibitors of integrin αVβ3 prevented these effects, similarly to the RGD-cyclic peptide compound Cilengitide, which suggests that the spike protein-through its RGD motif-binds to αVβ3 and elicits vascular leakage events. These findings support integrins as an additional receptor for SARS-CoV-2, particularly as integrin engagement can elucidate many of the adverse endothelial dysfunction events that stem from COVID-19.
Collapse
Affiliation(s)
| | - Steve W. Kerrigan
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences (PBS), RCSI University of Medicine and Health Sciences, 123 St. Stephen’s Green, D02 YN77 Dublin, Ireland;
| |
Collapse
|
14
|
The Effects of αvβ3 Integrin Blockage in Breast Tumor and Endothelial Cells under Hypoxia In Vitro. Int J Mol Sci 2022; 23:ijms23031745. [PMID: 35163668 PMCID: PMC8835904 DOI: 10.3390/ijms23031745] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is characterized by a hypoxic microenvironment inside the tumor mass, contributing to cell metastatic behavior. Hypoxia induces the expression of hypoxia-inducible factor (HIF-1α), a transcription factor for genes involved in angiogenesis and metastatic behavior, including the vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and integrins. Integrin receptors play a key role in cell adhesion and migration, being considered targets for metastasis prevention. We investigated the migratory behavior of hypoxia-cultured triple-negative breast cancer cells (TNBC) and endothelial cells (HUVEC) upon αvβ3 integrin blocking with DisBa-01, an RGD disintegrin with high affinity to this integrin. Boyden chamber, HUVEC transmigration, and wound healing assays in the presence of DisBa-01 were performed in hypoxic conditions. DisBa-01 produced similar effects in the two oxygen conditions in the Boyden chamber and transmigration assays. In the wound healing assay, hypoxia abolished DisBa-01′s inhibitory effect on cell motility and decreased the MMP-9 activity of conditioned media. These results indicate that αvβ3 integrin function in cell motility depends on the assay and oxygen levels, and higher inhibitor concentrations may be necessary to achieve the same inhibitory effect as in normoxia. These versatile responses add more complexity to the role of the αvβ3 integrin during tumor progression.
Collapse
|
15
|
Deshayes de Cambronne R, Fouet A, Picart A, Bourrel AS, Anjou C, Bouvier G, Candeias C, Bouaboud A, Costa L, Boulay AC, Cohen-Salmon M, Plu I, Rambaud C, Faurobert E, Albigès-Rizo C, Tazi A, Poyart C, Guignot J. CC17 group B Streptococcus exploits integrins for neonatal meningitis development. J Clin Invest 2021; 131:136737. [PMID: 33465054 DOI: 10.1172/jci136737] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
Group B Streptococcus (GBS) is the major cause of human neonatal infections. A single clone, designated CC17-GBS, accounts for more than 80% of meningitis cases, the most severe form of the infection. However, the events allowing blood-borne GBS to penetrate the brain remain largely elusive. In this study, we identified the host transmembrane receptors α5β1 and αvβ3 integrins as the ligands of Srr2, a major CC17-GBS-specific adhesin. Two motifs located in the binding region of Srr2 were responsible for the interaction between CC17-GBS and these integrins. We demonstrated in a blood-brain-barrier cellular model that both integrins contributed to the adhesion and internalization of CC17-GBS. Strikingly, both integrins were overexpressed during the postnatal period in the brain vessels of the blood-brain barrier and blood-cerebrospinal fluid barrier and contributed to juvenile susceptibility to CC17 meningitis. Finally, blocking these integrins decreased the ability of CC17-GBS to cross into the CNS of juvenile mice in an in vivo model of meningitis. Our study demonstrated that CC17-GBS exploits integrins in order to cross the brain vessels, leading to meningitis. Importantly, it provides host molecular insights into neonate's susceptibility to CC17-GBS meningitis, thereby opening new perspectives for therapeutic and prevention strategies of GBS-elicited meningitis.
Collapse
Affiliation(s)
| | - Agnès Fouet
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Amandine Picart
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Anne-Sophie Bourrel
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France.,Hôpitaux Universitaires Paris Centre, Cochin, Assistance Publique Hôpitaux de Paris, France
| | - Cyril Anjou
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Guillaume Bouvier
- Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, Institut Pasteur, CNRS UMR3528, C3BI, Paris, France
| | - Cristina Candeias
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Abdelouhab Bouaboud
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Lionel Costa
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Anne-Cécile Boulay
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, France
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, PSL Research University, Paris, France
| | - Isabelle Plu
- Sorbonne Université/Département de Neuropathologie Raymond Escourolle - Hôpital Pitié-Salpêtrière - Assistance Publique-Hôpitaux de Paris, France
| | - Caroline Rambaud
- Université de Versailles Saint Quentin en Yvelines (Université Paris-Saclay)/Service d'anatomie-pathologique et médecine légale, Hôpital Raymond Poincaré, Garches, France
| | - Eva Faurobert
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, France/Université Grenoble Alpes, La Tronche, France
| | - Corinne Albigès-Rizo
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, France/Université Grenoble Alpes, La Tronche, France
| | - Asmaa Tazi
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France.,Hôpitaux Universitaires Paris Centre, Cochin, Assistance Publique Hôpitaux de Paris, France.,Centre National de Référence des Streptocoques, France
| | - Claire Poyart
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France.,Hôpitaux Universitaires Paris Centre, Cochin, Assistance Publique Hôpitaux de Paris, France.,Centre National de Référence des Streptocoques, France
| | - Julie Guignot
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| |
Collapse
|
16
|
Amruta N, Bix G. ATN-161 Ameliorates Ischemia/Reperfusion-induced Oxidative Stress, Fibro-inflammation, Mitochondrial damage, and Apoptosis-mediated Tight Junction Disruption in bEnd.3 Cells. Inflammation 2021; 44:2377-2394. [PMID: 34420157 PMCID: PMC8380192 DOI: 10.1007/s10753-021-01509-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/25/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated the significance of endothelial cell-expressed α5β1 integrin in ischemic stroke, having shown that α5β1 integrin endothelial cell-selective knockout mice are significantly resistance to ischemic stroke injury via preservation of the tight junction protein claudin-5 and subsequent stabilization of the blood–brain barrier (BBB). In addition, inhibition of α5β1 by the small peptide noncompetitive integrin α5 inhibitor, ATN-161, is beneficial in a mouse model of ischemic stroke through reduction of infarct volume, edema, stabilization of the BBB, and reduced inflammation and immune cell infiltration into the brain. In continuation with our previous findings, we have further evaluated the mechanistic role of ATN-161 in vitro and found that oxygen and glucose deprivation and reperfusion (OGD/R)-induced inflammation, oxidative stress, apoptosis, mitochondrial depolarization, and fibrosis attenuate tight junction integrity via induction of α5, NLRP3, p-FAK, and p-AKT signaling in mouse brain endothelial cells. ATN-161 treatment (10 µM) effectively inhibited OGD/R-induced extracellular matrix (ECM) deposition by reducing integrin α5, MMP-9, and fibronectin expression, as well as reducing oxidative stress by reducing mitochondrial superoxide radicals, intracellular ROS, inflammation by reducing NLRP3 inflammasome, tight junction loss by reducing claudin-5 and ZO-1 expression levels, mitochondrial damage by inhibiting mitochondrial depolarization, and apoptosis via regulation of p-FAK and p-AKT levels. Taken together, our results further support therapeutically targeting α5 integrin with ATN-161, a safe, well-tolerated, and clinically validated peptide, in ischemic stroke.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA. .,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA. .,Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
| |
Collapse
|
17
|
SARS-CoV-2, Endothelial Dysfunction, and the Renin-Angiotensin System (RAS): A Potentially Dangerous Triad for the Development of Pre-Eclampsia. REPRODUCTIVE MEDICINE 2021. [DOI: 10.3390/reprodmed2020010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 represents the greatest epidemiological, clinical, and social challenge the human being has had to face in this century. SARS-CoV-2 is not merely a respiratory virus, as its target cells range from upper airway respiratory cells to pulmonary cells but also and above all to the cardiovascular cells, such as pericytes and endothelial cells. Indeed, the pathology related to SARS-CoV-2, COVID-19, may be defined as a thromboinflammatory syndrome in its most severe form, characterized by sepsis-induced coagulopathy (SIC) and disseminated intravascular coagulopathy (DIC), which is prevalent in individuals already presenting a chronic level of inflammation (e.g., obese individuals, elderly) and hypertension. Pregnancy is not only an inflammatory-prone condition but is characterized by a consistent rearrangement of the blood circulation and coagulation profile. Cardiac output increases while arterial systolic and diastolic pressure decrease, regardless of the activation of the RAS system. ACE2, the SARS-CoV-2 entry receptor into the host cells, which transforms Ang II in Ang 1–7, is highly expressed in endothelial, smooth muscle cells and pericytes of placental villi, regulating blood pressure and fetal development. Pre-eclampsia is a pregnancy disorder characterized by hypertension and low levels of ACE2, endothelial dysfunction, and a high production of pro-inflammatory cytokines, resembling COVID-19 manifestations. Whereas pre-eclampsia and COVID-19 have overlapping clinical features, a role for SARS-CoV-2 as a leading cause of pre-eclampsia in COVID-19 positive pregnant women has not been clarified yet. In this mini-review, we will explore the possibility of the existence of such a link, focusing on the role of endothelial dysfunction and RAS in both pre-eclampsia and SARS-CoV-2-induced COVID-19 pathogenesis.
Collapse
|
18
|
Pulous FE, Carnevale JC, Al-Yafeai Z, Pearson BH, Hamilton JAG, Henry CJ, Orr AW, Petrich BG. Talin-dependent integrin activation is required for endothelial proliferation and postnatal angiogenesis. Angiogenesis 2021; 24:177-190. [PMID: 33113074 PMCID: PMC8441968 DOI: 10.1007/s10456-020-09756-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
Integrin activation contributes to key blood cell functions including adhesion, proliferation and migration. An essential step in the cell signaling pathway that activates integrin requires the binding of talin to the β-integrin cytoplasmic tail. Whereas this pathway is understood in platelets in detail, considerably less is known regarding how integrin-mediated adhesion in endothelium contributes to postnatal angiogenesis. We utilized an inducible EC-specific talin1 knock-out mouse (Tln1 EC-KO) and talin1 L325R knock-in mutant (Tln1 L325R) mouse, in which talin selectively lacks the capacity to activate integrins, to assess the role of integrin activation during angiogenesis. Deletion of talin1 during postnatal days 1-3 (P1-P3) caused lethality by P8 with extensive defects in retinal angiogenesis and widespread hemorrhaging. Tln1 EC-KO mice displayed reduced retinal vascular area, impaired EC sprouting and proliferation relative to Tln1 CTRLs. In contrast, induction of talin1 L325R in neonatal mice resulted in modest defects in retinal angiogenesis and mice survived to adulthood. Interestingly, deletion of talin1 or expression of talin1 L325R in ECs increased MAPK/ERK signaling. Strikingly, B16-F0 tumors grown in Tln1 L325R adult mice were 55% smaller and significantly less vascularized than tumors grown in littermate controls. EC talin1 is indispensable for postnatal development angiogenesis. The role of EC integrin activation appears context-dependent as its inhibition is compatible with postnatal development with mild defects in retinal angiogenesis but results in marked defects in tumor growth and angiogenesis. Inhibiting EC pan-integrin activation may be an effective approach to selectively target tumor blood vessel growth.
Collapse
Affiliation(s)
- Fadi E Pulous
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Jamie C Carnevale
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Zaki Al-Yafeai
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Brenna H Pearson
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Jamie A G Hamilton
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Curtis J Henry
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
- Department of Cell Biology and Anatomy, LSU Health Sciences Center, Shreveport, LA, USA
- Pathology and Translational Pathobiology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Brian G Petrich
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
19
|
Matjuda EN, Engwa GA, Sewani-Rusike CR, Nkeh-Chungag BN. An Overview of Vascular Dysfunction and Determinants: The Case of Children of African Ancestry. Front Pediatr 2021; 9:769589. [PMID: 34956981 PMCID: PMC8709476 DOI: 10.3389/fped.2021.769589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
The balance between dilatory and constrictive factors is important as it keeps blood vessels in a homeostatic state. However, altered physiological processes as a result of obesity, hypertension, oxidative stress, and other cardiovascular risk factors may lead to vascular damage, causing an imbalance of vasoactive factors. Over time, the sustained imbalance of these vasoactive factors may lead to vascular dysfunction, which can be assessed by non-invasive methods, such as flow-mediated dilation, pulse wave velocity, flow-mediated slowing, retinal vessel analysis, peripheral vascular reactivity, and carotid intima-media thickness assessment. Although there is increasing prevalence of cardiovascular risk factors (obesity and hypertension) in children in sub-Saharan Africa, little is known about how this may affect vascular function. This review focuses on vasoactive factors implicated in vascular (dys)function, highlighting the determinants and consequences of vascular dysfunction. It further describes the non-invasive methods used for vascular (dys)function assessments and, last, describes the impact of cardiovascular risk factors on vascular dysfunction in children of African ancestry.
Collapse
Affiliation(s)
- Edna N Matjuda
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Godwill Azeh Engwa
- Department of Biological and Environmental Sciences, Faculty of Natural Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Constance R Sewani-Rusike
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Benedicta N Nkeh-Chungag
- Department of Biological and Environmental Sciences, Faculty of Natural Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| |
Collapse
|
20
|
Ristori E, Cicaloni V, Salvini L, Tinti L, Tinti C, Simons M, Corti F, Donnini S, Ziche M. Amyloid-β Precursor Protein APP Down-Regulation Alters Actin Cytoskeleton-Interacting Proteins in Endothelial Cells. Cells 2020; 9:cells9112506. [PMID: 33228083 PMCID: PMC7699411 DOI: 10.3390/cells9112506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous membrane protein often associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Despite its role in the development of the pathogenesis, APP exerts several physiological roles that have been mainly investigated in neuronal tissue. To date, the role of APP in vasculature and endothelial cells has not been fully elucidated. In this study, we used molecular and proteomic approaches to identify and investigate major cellular targets of APP down-regulation in endothelial cells. We found that APP is necessary for endothelial cells proliferation, migration and adhesion. The loss of APP alters focal adhesion stability and cell-cell junctions' expression. Moreover, APP is necessary to mediate endothelial response to the VEGF-A growth factor. Finally, we document that APP propagates exogenous stimuli and mediates cellular response in endothelial cells by modulating the Scr/FAK signaling pathway. Thus, the intact expression and processing of APP is required for normal endothelial function. The identification of molecular mechanisms responsible for vasoprotective properties of endothelial APP may have an impact on clinical efforts to preserve and protect healthy vasculature in patients at risk of the development of cerebrovascular disease and dementia including AD and CAA.
Collapse
Affiliation(s)
- Emma Ristori
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Vittoria Cicaloni
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Salvini
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Cristina Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Michael Simons
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
- Departments of Medicine (Cardiology) and Cell Biology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Federico Corti
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
| | - Sandra Donnini
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| | - Marina Ziche
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| |
Collapse
|
21
|
Kim DH, Ahn J, Kang HK, Kim MS, Kim NG, Kook MG, Choi SW, Jeon NL, Woo HM, Kang KS. Development of highly functional bioengineered human liver with perfusable vasculature. Biomaterials 2020; 265:120417. [PMID: 32987272 DOI: 10.1016/j.biomaterials.2020.120417] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/28/2020] [Accepted: 09/19/2020] [Indexed: 12/11/2022]
Abstract
Liver tissue engineering offers a promising strategy for liver failure patients. Since transplantation rejection resulting in vessel thrombosis is regarded as a major hurdle, vascular reconstruction is one of indispensable requirements of whole organ engineering. Here we demonstrated a novel strategy for reconstruction of a vascularized bioengineered human liver (VBHL) using decellularized liver scaffolds in an efficient manner. First we achieved fully functional endothelial coverage of scaffolds by adopting the anti-CD31 aptamer as a potent coating agent for re-endothelialization. Through an ex vivo human blood perfusion that recapitulates the blood coagulation response in humans, we demonstrated significantly reduced platelet aggregation in anti-CD31 aptamer coated scaffolds. We then produced VBHL constructs using liver parenchymal cells and nonparenchymal cells, properly organized into liver-like structures with an aligned vasculature. Interestingly, VBHL constructs displayed prominently enhanced long-term liver-specific functions that are affected by vascular functionality. The VBHL constructs formed perfusable vessel networks in vivo as evidenced by the direct vascular connection between the VBHL constructs and the renal circulation. Furthermore, heterotopic transplantation of VBHL constructs supported liver functions in a rat model of liver fibrosis. Overall, we proposed a new strategy to generate transplantable bioengineered livers characterized by highly functional vascular reconstruction.
Collapse
Affiliation(s)
- Da-Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jungho Ahn
- School of Mechanical Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Hyun Kyoung Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Soo Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Nam-Gyo Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Myung Geun Kook
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Noo Li Jeon
- School of Mechanical Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Heung-Myong Woo
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Botros L, Pronk MCA, Juschten J, Liddle J, Morsing SKH, van Buul JD, Bates RH, Tuinman PR, van Bezu JSM, Huveneers S, Bogaard HJ, van Hinsbergh VWM, Hordijk PL, Aman J. Bosutinib prevents vascular leakage by reducing focal adhesion turnover and reinforcing junctional integrity. J Cell Sci 2020; 133:jcs240077. [PMID: 32198280 DOI: 10.1242/jcs.240077] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/11/2020] [Indexed: 12/29/2022] Open
Abstract
Endothelial barrier dysfunction leads to edema and vascular leak, causing high morbidity and mortality. Previously, Abl kinase inhibition has been shown to protect against vascular leak. Using the distinct inhibitory profiles of clinically available Abl kinase inhibitors, we aimed to provide a mechanistic basis for novel treatment strategies against vascular leakage syndromes. We found that the inhibitor bosutinib most potently protected against inflammation-induced endothelial barrier disruption. In vivo, bosutinib prevented lipopolysaccharide (LPS)-induced alveolar protein extravasation in an acute lung injury mice model. Mechanistically, mitogen-activated protein 4 kinase 4 (MAP4K4) was identified as important novel mediator of endothelial permeability, which signaled via ezrin, radixin and moesin proteins to increase turnover of integrin-based focal adhesions. The combined inhibition of MAP4K4 and Abl-related gene (Arg, also known as ABL2) by bosutinib preserved adherens junction integrity and reduced turnover of focal adhesions, which synergistically act to stabilize the endothelial barrier during inflammation. We conclude that MAP4K4 is an important regulator of endothelial barrier integrity, increasing focal adhesion turnover and disruption of cell-cell junctions during inflammation. Because it inhibits both Arg and MAP4K4, use of the clinically available drug bosutinib might form a viable strategy against vascular leakage syndromes.
Collapse
Affiliation(s)
- Liza Botros
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Manon C A Pronk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Jenny Juschten
- Amsterdam UMC, University of Amsterdam, Department of Intensive Care, 1105 AZ Amsterdam, The Netherlands
| | - John Liddle
- GlaxoSmithKline, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Sofia K H Morsing
- Molecular Cell Biology Lab at Dept. Molecular Cellular Haemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Dept. Molecular Cellular Haemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | | | - Pieter R Tuinman
- Amsterdam UMC, University of Amsterdam, Department of Intensive Care, 1105 AZ Amsterdam, The Netherlands
| | - Jan S M van Bezu
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Victor W M van Hinsbergh
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Peter L Hordijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| | - Jurjan Aman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonology, Amsterdam Cardiovascular Sciences, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|