1
|
Lemmens TP, Bröker V, Rijpkema M, Hughes CCW, Schurgers LJ, Cosemans JMEM. Fundamental considerations for designing endothelialized in vitro models of thrombosis. Thromb Res 2024; 236:179-190. [PMID: 38460307 DOI: 10.1016/j.thromres.2024.03.004] [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: 11/18/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Endothelialized in vitro models for cardiovascular disease have contributed greatly to our current understanding of the complex molecular mechanisms underlying thrombosis. To further elucidate these mechanisms, it is important to consider which fundamental aspects to incorporate into an in vitro model. In this review, we will focus on the design of in vitro endothelialized models of thrombosis. Expanding our understanding of the relation and interplay between the different pathways involved will rely in part on complex models that incorporate endothelial cells, blood, the extracellular matrix, and flow. Importantly, the use of tissue-specific endothelial cells will help in understanding the heterogeneity in thrombotic responses between different vascular beds. The dynamic and complex responses of endothelial cells to different shear rates underlines the importance of incorporating appropriate shear in in vitro models. Alterations in vascular extracellular matrix composition, availability of bioactive molecules, and gradients in concentration and composition of these molecules can all regulate the function of both endothelial cells and perivascular cells. Factors modulating these elements in in vitro models should therefore be considered carefully depending on the research question at hand. As the complexity of in vitro models increases, so can the variability. A bottom-up approach to designing such models will remain an important tool for researchers studying thrombosis. As new techniques are continuously being developed and new pathways are brought to light, research question-dependent considerations will have to be made regarding what aspects of thrombosis to include in in vitro models.
Collapse
Affiliation(s)
- Titus P Lemmens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Vanessa Bröker
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Minke Rijpkema
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, and Department of Biomedical Engineering, University of California, Irvine, USA
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
2
|
Santiago FS, Li Y, Khachigian LM. Serine 26 in Early Growth Response-1 Is Critical for Endothelial Proliferation, Migration, and Network Formation. J Am Heart Assoc 2021; 10:e020521. [PMID: 34476983 PMCID: PMC8649526 DOI: 10.1161/jaha.120.020521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Vascular endothelial cell proliferation, migration, and network formation are key proangiogenic processes involving the prototypic immediate early gene product, Egr‐1 (early growth response‐1). Egr‐1 undergoes phosphorylation at a conserved Ser26 but its function is completely unknown in endothelial cells or any other cell type. Methods and Results A CRISPR/Cas9 strategy was used to introduce a homozygous Ser26>Ala mutation into endogenous Egr‐1 in human microvascular endothelial cells. In the course of generating mutant cells, we produced cells with homozygous deletion in Egr‐1 caused by frameshift and premature termination. We found that Ser26 mutation in Egr‐1, or Egr‐1 deletion, perturbed endothelial cell proliferation in models of cell counting or real‐time growth using the xCELLigence System. We found that Ser26 mutation or Egr‐1 deletion ameliorated endothelial cell migration toward VEGF‐A165 (vascular endothelial growth factor‐A) in a dual‐chamber model. On solubilized basement membrane preparations, Ser26 mutation or Egr‐1 deletion prevented endothelial network (or tubule) formation, an in vitro model of angiogenesis. Flow cytometry further revealed that Ser26 mutation or Egr‐1 deletion elevated early and late apoptosis. Finally, we demonstrated that Ser26 mutation or Egr‐1 deletion increased VE‐cadherin (vascular endothelial cadherin) expression, a regulator of endothelial adhesion and signaling, permeability, and angiogenesis. Conclusions These findings not only indicate that Egr‐1 is essential for endothelial cell proliferation, migration, and network formation, but also show that point mutation in Ser26 is sufficient to impair each of these processes and trigger apoptosis as effectively as the absence of Egr‐1. This highlights the importance of Ser26 in Egr‐1 for a range of proangiogenic processes.
Collapse
Affiliation(s)
- Fernando S Santiago
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Yue Li
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| |
Collapse
|
3
|
Kedem A, Aelion-Brauer A, Guo P, Wen D, Ding BS, Lis R, Cheng D, Sandler VM, Rafii S, Rosenwaks Z. Activated ovarian endothelial cells promote early follicular development and survival. J Ovarian Res 2017; 10:64. [PMID: 28927438 PMCID: PMC5605973 DOI: 10.1186/s13048-017-0354-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/31/2017] [Indexed: 12/30/2022] Open
Abstract
Background New data suggests that endothelial cells (ECs) elaborate essential “angiocrine factors”. The aim of this study is to investigate the role of activated ovarian endothelial cells in early in-vitro follicular development. Methods Mouse ovarian ECs were isolated using magnetic cell sorting or by FACS and cultured in serum free media. After a constitutive activation of the Akt pathway was initiated, early follicles (50–150 um) were mechanically isolated from 8-day-old mice and co-cultured with these activated ovarian endothelial cells (AOEC) (n = 32), gel (n = 24) or within matrigel (n = 27) in serum free media for 14 days. Follicular growth, survival and function were assessed. Results After 6 passages, flow cytometry showed 93% of cells grown in serum-free culture were VE-cadherin positive, CD-31 positive and CD 45 negative, matching the known EC profile. Beginning on day 4 of culture, we observed significantly higher follicular and oocyte growth rates in follicles co-cultured with AOECs compared with follicles on gel or matrigel. After 14 days of culture, 73% of primary follicles and 83% of secondary follicles co-cultured with AOEC survived, whereas the majority of follicles cultured on gel or matrigel underwent atresia. Conclusions This is the first report of successful isolation and culture of ovarian ECs. We suggest that co-culture with activated ovarian ECs promotes early follicular development and survival. This model is a novel platform for the in vitro maturation of early follicles and for the future exploration of endothelial-follicular communication. Capsule In vitro development of early follicles necessitates a complex interplay of growth factors and signals required for development. Endothelial cells (ECs) may elaborate essential “angiocrine factors” involved in organ regeneration. We demonstrate that co-culture with ovarian ECs enables culture of primary and early secondary mouse ovarian follicles.
Collapse
Affiliation(s)
- Alon Kedem
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA. .,Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA.
| | - Anate Aelion-Brauer
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA.
| | - Peipei Guo
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Duancheng Wen
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Bi-Sen Ding
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Raphael Lis
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Du Cheng
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Vladislav M Sandler
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Shahin Rafii
- Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| | - Zev Rosenwaks
- The Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10021, USA.,Ansary Stem Cell Center for Regenerative Medicine at Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
4
|
Schlinkert P, Casals E, Boyles M, Tischler U, Hornig E, Tran N, Zhao J, Himly M, Riediker M, Oostingh GJ, Puntes V, Duschl A. The oxidative potential of differently charged silver and gold nanoparticles on three human lung epithelial cell types. J Nanobiotechnology 2015; 13:1. [PMID: 25592092 PMCID: PMC4304186 DOI: 10.1186/s12951-014-0062-4] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/18/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticle (NPs) functionalization has been shown to affect their cellular toxicity. To study this, differently functionalized silver (Ag) and gold (Au) NPs were synthesised, characterised and tested using lung epithelial cell systems. METHODS Monodispersed Ag and Au NPs with a size range of 7 to 10 nm were coated with either sodium citrate or chitosan resulting in surface charges from -50 mV to +70 mV. NP-induced cytotoxicity and oxidative stress were determined using A549 cells, BEAS-2B cells and primary lung epithelial cells (NHBE cells). TEER measurements and immunofluorescence staining of tight junctions were performed to test the growth characteristics of the cells. Cytotoxicity was measured by means of the CellTiter-Blue ® and the lactate dehydrogenase assay and cellular and cell-free reactive oxygen species (ROS) production was measured using the DCFH-DA assay. RESULTS Different growth characteristics were shown in the three cell types used. A549 cells grew into a confluent mono-layer, BEAS-2B cells grew into a multilayer and NHBE cells did not form a confluent layer. A549 cells were least susceptible towards NPs, irrespective of the NP functionalization. Cytotoxicity in BEAS-2B cells increased when exposed to high positive charged (+65-75 mV) Au NPs. The greatest cytotoxicity was observed in NHBE cells, where both Ag and Au NPs with a charge above +40 mV induced cytotoxicity. ROS production was most prominent in A549 cells where Au NPs (+65-75 mV) induced the highest amount of ROS. In addition, cell-free ROS measurements showed a significant increase in ROS production with an increase in chitosan coating. CONCLUSIONS Chitosan functionalization of NPs, with resultant high surface charges plays an important role in NP-toxicity. Au NPs, which have been shown to be inert and often non-cytotoxic, can become toxic upon coating with certain charged molecules. Notably, these effects are dependent on the core material of the particle, the cell type used for testing and the growth characteristics of these cell culture model systems.
Collapse
Affiliation(s)
- Paul Schlinkert
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| | - Eudald Casals
- Institute Catalá de Nanotecnologia, Barcelona, Spain.
| | - Matthew Boyles
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| | - Ulrike Tischler
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| | - Eva Hornig
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| | - Ngoc Tran
- Institute Catalá de Nanotecnologia, Barcelona, Spain.
| | - Jiayuan Zhao
- Institute for Work and Health, Lausanne, Switzerland.
| | - Martin Himly
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| | - Michael Riediker
- Institute for Work and Health, Lausanne, Switzerland.
- Institue for Occupational Medicine (IOM) Singapore, Downtown Core, Singapore.
| | - Gertie Janneke Oostingh
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
- Biomedical Sciences, Salzburg University of Applied Sciences, Puch, Salzburg, Austria.
| | - Victor Puntes
- Institute Catalá de Nanotecnologia, Barcelona, Spain.
| | - Albert Duschl
- Department of Molecular Biology, Paris Lodron-University of Salzburg, Hellbrunnerstr. 34, A-5020, Salzburg, Austria.
| |
Collapse
|
5
|
Forkel S, Schön M, Hildmann A, Claßen A, John SM, Danker K, Schön MP. Inositoylated platelet-activating factor (Ino-C2-PAF) modulates dynamic lymphocyte-endothelial cell interactions and alleviates psoriasis-like skin inflammation in two complementary mouse models. J Invest Dermatol 2014; 134:2510-2520. [PMID: 24714204 DOI: 10.1038/jid.2014.170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/23/2014] [Accepted: 03/14/2014] [Indexed: 02/08/2023]
Abstract
Psoriasis, a tumor necrosis factor alpha (TNFα)-governed inflammatory disorder with prominent dysregulation of cutaneous vascular functions, has evolved into a model disorder for studying anti-inflammatory therapies. We present experimental in vitro and in vivo data on 1-O-octadecyl-2-O-(2-(myo-inositolyl)-ethyl)-sn-glycero-3-(R/S)-phosphatidyl-choline (Ino-C2-PAF), the lead compound of a class of synthetic glycosylated phospholipids, in anti-inflammatory therapy. Ino-C2-PAF strongly induced apoptosis only in TNFα-stimulated, but not in untreated human vascular endothelial cells. Moreover, TNFα-induced endothelial adhesion molecules that mediated the rolling and firm adhesion of leukocytes (vascular cell adhesion protein-1 (VCAM-1), E-selectin, and ICAM-1) were selectively downregulated by Ino-C2-PAF. Similarly, expression of L-selectin, VCAM-1 receptor α4β1 integrin , and lymphocyte function-associated antigen-1 on human peripheral blood mononuclear cells was reduced without induction of apoptosis. Functionally, these changes were accompanied by significant impairment of rolling and adhesion of human peripheral blood lymphocytes on TNFα-activated endothelial cells in a dynamic flow chamber system. When the therapeutic potential of Ino-C2-PAF was assessed in two complementary mouse models of psoriasis, K5.hTGFβ1 transgenic and JunB/c-Jun-deficient mice, Ino-C2-PAF led to significant alleviation of the clinical symptoms and normalized the pathological cutaneous changes including vascularization. There were no overt adverse effects. These findings suggested that Ino-C2-PAF is a potential candidate in the therapy of inflammatory skin diseases that include abnormal vascular functions.
Collapse
Affiliation(s)
- Susann Forkel
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | - Margarete Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | - Annette Hildmann
- Institute of Biochemistry, Charité University Medical Center, Berlin, Germany
| | - Anna Claßen
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany
| | - Swen-Malte John
- Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany; Department of Dermatology, Environmental Medicine and Health Care Theory, University of Osnabrück, Osnabrück, Germany
| | - Kerstin Danker
- Institute of Biochemistry, Charité University Medical Center, Berlin, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany.
| |
Collapse
|
6
|
Lockmann A, Schön MP. TNFα-induced leukocyte-endothelial cell interactions show marked interindividual differences independent of the clinical response to adalimumab. Exp Dermatol 2014; 23:133-4. [DOI: 10.1111/exd.12283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Anike Lockmann
- Department of Dermatology, Venereology and Allergology; University Medical Center; Georg August University; Göttingen Germany
| | - Michael P. Schön
- Department of Dermatology, Venereology and Allergology; University Medical Center; Georg August University; Göttingen Germany
| |
Collapse
|
7
|
Keuschnigg J, Karinen S, Auvinen K, Irjala H, Mpindi JP, Kallioniemi O, Hautaniemi S, Jalkanen S, Salmi M. Plasticity of blood- and lymphatic endothelial cells and marker identification. PLoS One 2013; 8:e74293. [PMID: 24058540 PMCID: PMC3769239 DOI: 10.1371/journal.pone.0074293] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/29/2013] [Indexed: 01/17/2023] Open
Abstract
The distinction between lymphatic and blood vessels is biologically fundamental. Here we wanted to rigorously analyze the universal applicability of vascular markers and characteristics of the two widely used vascular model systems human microvascular endothelial cell line-1 (HMEC-1) and telomerase-immortalized microvascular endothelial cell line (TIME). Therefore we studied the protein expression and functional properties of the endothelial cell lines HMEC-1 and TIME by flow cytometry and in vitro flow assays. We then performed microarray analyses of the gene expression in these two cell lines and compared them to primary endothelial cells. Using bioinformatics we then defined 39 new, more universal, endothelial-type specific markers from 47 primary endothelial microarray datasets and validated them using immunohistochemistry with normal and pathological tissues. We surprisingly found that both HMEC-1 and TIME are hybrid blood- and lymphatic cells. In addition, we discovered great discrepancies in the previous identifications of blood- and lymphatic endothelium-specific genes. Hence we identified and validated new, universally applicable vascular markers. Summarizing, the hybrid blood-lymphatic endothelial phenotype of HMEC-1 and TIME is indicative of plasticity in the gene expression of immortalized endothelial cell lines. Moreover, we identified new, stable, vessel-type specific markers for blood- and lymphatic endothelium, useful for basic research and clinical diagnostics.
Collapse
Affiliation(s)
- Johannes Keuschnigg
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
- Turku Doctoral Program of Biomedical Sciences, Turku, Finland
| | - Sirkku Karinen
- Research Programs Unit, Genome-Scale Biology, and Institute of Biomedicine, Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland
- National Institute of Public Health and Welfare, Turku, Finland
| | - Heikki Irjala
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - John-Patrick Mpindi
- FIMM, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- FIMM, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology, and Institute of Biomedicine, Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
- National Institute of Public Health and Welfare, Turku, Finland
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland
- National Institute of Public Health and Welfare, Turku, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
- * E-mail:
| |
Collapse
|
8
|
Lockmann A, Schön MP. Phenotypic and functional traits of peripheral blood mononuclear cells retained by controlled cryopreservation: implications for reliable sequential studies of dynamic interactions with endothelial cells. Exp Dermatol 2013; 22:358-9. [DOI: 10.1111/exd.12123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Anike Lockmann
- Department of Dermatology, Venereology and Allergology; University Medical Center, Georg August University; Göttingen Germany
| | - Michael P. Schön
- Department of Dermatology, Venereology and Allergology; University Medical Center, Georg August University; Göttingen Germany
| |
Collapse
|
9
|
Seidl K, Solis NV, Bayer AS, Hady WA, Ellison S, Klashman MC, Xiong YQ, Filler SG. Divergent responses of different endothelial cell types to infection with Candida albicans and Staphylococcus aureus. PLoS One 2012; 7:e39633. [PMID: 22745797 PMCID: PMC3382135 DOI: 10.1371/journal.pone.0039633] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/27/2012] [Indexed: 01/18/2023] Open
Abstract
Endothelial cells are important in the pathogenesis of bloodstream infections caused by Candida albicans and Staphylococcus aureus. Numerous investigations have used human umbilical vein endothelial cells (HUVECs) to study microbial-endothelial cell interactions in vitro. However, the use of HUVECs requires a constant supply of umbilical cords, and there are significant donor-to-donor variations in these endothelial cells. The use of an immortalized endothelial cell line would obviate such difficulties. One candidate in this regard is HMEC-1, an immortalized human dermal microvascular endothelial cell line. To determine if HMEC-1 cells are suitable for studying the interactions of C. albicans and S. aureus with endothelial cells in vitro, we compared the interactions of these organisms with HMEC-1 cells and HUVECs. We found that wild-type C. albicans had significantly reduced adherence to and invasion of HMEC-1 cells as compared to HUVECs. Although wild-type S. aureus adhered to and invaded HMEC-1 cells similarly to HUVECs, an agr mutant strain had significantly reduced invasion of HMEC-1 cells, but not HUVECs. Furthermore, HMEC-1 cells were less susceptible to damage induced by C. albicans, but more susceptible to damage caused by S. aureus. In addition, HMEC-1 cells secreted very little IL-8 in response to infection with either organism, whereas infection of HUVECs induced substantial IL-8 secretion. This weak IL-8 response was likely due to the anatomic site from which HMEC-1 cells were obtained because infection of primary human dermal microvascular endothelial cells with C. albicans and S. aureus also induced little increase in IL-8 production above basal levels. Thus, C. albicans and S. aureus interact with HMEC-1 cells in a substantially different manner than with HUVECs, and data obtained with one type of endothelial cell cannot necessarily be extrapolated to other types.
Collapse
Affiliation(s)
- Kati Seidl
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Norma V. Solis
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Arnold S. Bayer
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Wessam Abdel Hady
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Steven Ellison
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- Department of Biology, California State University-Dominguez Hills, Carson, California, United States of America
| | - Meredith C. Klashman
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Yan Q. Xiong
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Scott G. Filler
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
10
|
Wallbrecht K, Drick N, Hund AC, Schön MP. Downregulation of endothelial adhesion molecules by dimethylfumarate, but not monomethylfumarate, and impairment of dynamic lymphocyte-endothelial cell interactions. Exp Dermatol 2011; 20:980-5. [PMID: 21995308 DOI: 10.1111/j.1600-0625.2011.01376.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although fumaric acid esters (FAE) have a decade-long firm place in the therapeutic armamentarium for psoriasis, their pleiotropic mode of action is not yet fully understood. While most previous studies have focused on the effects of FAE on leucocytes, we have addressed their activity on macro- and microvascular endothelial cells. As detected both on mRNA and protein levels, dimethylfumarate effected a profound reduction of TNFα-induced expression of E-selectin (CD62E), ICAM-1 (CD54) and VCAM-1 (CD106) on two different endothelial cell populations in a concentration-dependent manner. This reduction of several endothelial adhesion molecules was accompanied by a dramatic diminution of both rolling and firm adhesive interactions between endothelial cells and lymphocytes in a dynamic flow chamber system. Dimethylfumarate, at a concentration of 50 μm, reduced lymphocyte rolling on endothelial cells by 85.9% (P<0.001 compared to untreated controls), and it diminished the number of adherent cells by 88% (P<0.001). In contrast, monomethylfumarate (MMF) influenced neither surface expression of adhesion molecules nor interactions between endothelial cells and lymphocytes. These observations demonstrate that endothelial cells, in addition to the known effects on leucocytes, undergo profound functional changes in response to dimethylfumarate. These changes are accompanied by severely impaired dynamic interactions with lymphocytes, which constitute the critical initial step of leucocyte recruitment to inflamed tissues in psoriasis and other TNF-related inflammatory disorders.
Collapse
Affiliation(s)
- Katrin Wallbrecht
- Department of Dermatology, Venereology and Allergology, Georg August University, Göttingen, Germany
| | | | | | | |
Collapse
|
11
|
Ferrero MC, Bregante J, Delpino MV, Barrionuevo P, Fossati CA, Giambartolomei GH, Baldi PC. Proinflammatory response of human endothelial cells to Brucella infection. Microbes Infect 2011; 13:852-61. [PMID: 21621633 DOI: 10.1016/j.micinf.2011.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 04/18/2011] [Accepted: 04/19/2011] [Indexed: 01/18/2023]
Abstract
Although vascular pathologies such as vasculitis, endocarditis and mycotic aneurysms have been described in brucellosis patients, the interaction of Brucella with the endothelium has not been characterized. In this study we show that Brucella abortus and Brucella suis can infect and replicate in primary human umbilical vein endothelial cells (HUVEC) and in the microvascular endothelial cell line HMEC-1. Infection led to an increased production of IL-8, MCP-1 and IL-6 in HUVEC and HMEC-1 cells, and an increased expression of adhesion molecules (CD54 in both cells, CD106 and CD62E in HUVEC). Experiments with purified antigens from the bacterial outer membrane revealed that lipoproteins (Omp19) but not lipopolysaccharide mediate these proinflammatory responses. Infection of polarized HMEC-1 cells resulted in an increased capacity of these cells to promote the transmigration of neutrophils from the apical to the basolateral side of the monolayer, and the same phenomenon was observed when the cells were stimulated with live bacteria from the basolateral side. Overall, these results suggest that Brucella spp. can infect and survive within endothelial cells, and can induce a proinflammatory response that might be involved in the vascular manifestations of brucellosis.
Collapse
Affiliation(s)
- Mariana C Ferrero
- Instituto de Estudios de la Inmunidad Humoral, Facultad de Farmacia y Bioquímica, UBA, Junín 956, (1113) Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
12
|
Steiner O, Coisne C, Engelhardt B, Lyck R. Comparison of immortalized bEnd5 and primary mouse brain microvascular endothelial cells as in vitro blood-brain barrier models for the study of T cell extravasation. J Cereb Blood Flow Metab 2011; 31:315-27. [PMID: 20606687 PMCID: PMC3049495 DOI: 10.1038/jcbfm.2010.96] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Important insights into the molecular mechanism of T cell extravasation across the blood-brain barrier (BBB) have already been obtained using immortalized mouse brain endothelioma cell lines (bEnd). However, compared with bEnd, primary brain endothelial cells have been shown to establish better barrier characteristics, including complex tight junctions and low permeability. In this study, we asked whether bEnd5 and primary mouse brain microvascular endothelial cells (pMBMECs) were equally suited as in vitro models with which to study the cellular and molecular mechanisms of T cell extravasation across the BBB. We found that both in vitro BBB models equally supported both T cell adhesion under static and physiologic flow conditions, and T cell crawling on the endothelial surface against the direction of flow. In contrast, distances of T cell crawling on pMBMECs were strikingly longer than on bEnd5, whereas diapedesis of T cells across pMBMECs was dramatically reduced compared with bEnd5. Thus, both in vitro BBB models are suited to study T cell adhesion. However, because pMBMECs better reflect endothelial BBB specialization in vivo, we propose that more reliable information about the cellular and molecular mechanisms of T cell diapedesis across the BBB can be attained using pMBMECs.
Collapse
Affiliation(s)
- Oliver Steiner
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | | | |
Collapse
|
13
|
Paclitaxel potentiates inflammatory cytokine-induced prothrombotic molecules in endothelial cells. J Cardiovasc Pharmacol 2010; 55:276-85. [PMID: 20075745 DOI: 10.1097/fjc.0b013e3181d263f7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
To overcome the limitations of balloon expandible metal stent-induced neointimal smooth muscle cell proliferation, drug-coated stent devices have been developed. Drug eluting stents release high concentrations of antiproliferative agents, such as paclitaxel, to reduce neointimal hyperplasia. The proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), is known to cause severe endothelial dysfunction and accelerate atherosclerotic lesion progression. The interaction of TNF-alpha and paclitaxel on the release of prothrombotic molecules was examined in endothelial cells. Treatment of endothelial cells with paclitaxel had no direct effect on tissue factor (TF) expression, but TNF-alpha increased TF. Cotreatment of paclitaxel with TNF-alpha markedly augmented the release of TF. TNF-alpha induced release of plasminogen activator inhibitor but no synergism occurred with paclitaxel. Treatment of endothelial cells with paclitaxel and TNF-alpha reduced expression of thrombomodulin and protein C receptor. Tissue factor pathway inhibitor expression was reduced by prolonged treatment with either paclitaxel or TNF-alpha. The adhesion molecule, CD62 E, was induced by TNF-alpha; however, CD31, CD62 P, and CD106 were not affected by paclitaxel and TNF-alpha. Apoptosis was not observed with cotreatment of endothelial cells with paclitaxel and TNF-alpha. CD59-positive microparticles were released in response to TNF-alpha, but the release was not augmented by paclitaxel. Paclitaxel and TNF-alpha increased the nitrotyrosination of proteins. These findings indicate that paclitaxel enhances TNF-alpha-induced release of TF, and downregulated thrombomodulin, increased protein nitration, which may subsequently favor prothrombotic intimal surface.
Collapse
|
14
|
Butler JM, Kobayashi H, Rafii S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat Rev Cancer 2010; 10:138-46. [PMID: 20094048 PMCID: PMC2944775 DOI: 10.1038/nrc2791] [Citation(s) in RCA: 433] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The precise mechanisms whereby anti-angiogenesis therapy blocks tumour growth or causes vascular toxicity are unknown. We propose that endothelial cells establish a vascular niche that promotes tumour growth and tissue repair not only by delivering nutrients and O2 but also through an 'angiocrine' mechanism by producing stem and progenitor cell-active trophogens. Identification of endothelial-derived instructive angiocrine factors will allow direct tumour targeting, while diminishing the unwanted side effects associated with the use of anti-angiogenic agents.
Collapse
Affiliation(s)
- Jason M Butler
- Hideki Kobayashi and Shahin Rafii are at the Howard Hughes Medical Institute, Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
15
|
Amschler K, Schön MP, Pletz N, Wallbrecht K, Erpenbeck L, Schön M. NF-kappaB inhibition through proteasome inhibition or IKKbeta blockade increases the susceptibility of melanoma cells to cytostatic treatment through distinct pathways. J Invest Dermatol 2009; 130:1073-86. [PMID: 19940859 DOI: 10.1038/jid.2009.365] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metastasized melanoma is almost universally resistant to chemotherapy. Given that constitutive or drug-induced upregulation of NF-kappaB activity is associated with this chemoresistance, NF-kappaB inhibition may increase the susceptibility to antitumoral therapy. On the cellular level, two principles of NF-kappaB inhibition, proteasome inhibition by bortezomib and IkappaB kinase-beta (IKKbeta) inhibition by the kinase inhibitor of NF-kappaB-1 (KINK-1), significantly increased the antitumoral efficacy of camptothecin. When combined with camptothecin, either of the two NF-kappaB-inhibiting principles synergistically influenced progression-related in vitro functions, including cell growth, apoptosis, and invasion through an artificial basement membrane. In addition, when C57BL/6 mice were intravenously injected with B16F10 melanoma cells, the combination of cytostatic treatment with either of the NF-kappaB-inhibiting compounds revealed significantly reduced pulmonary metastasis compared to either treatment alone. However, on the molecular level, nuclear translocation of p65, cell cycle analysis, and expression of NF-kappaB-dependent gene products disclosed distinctly different molecular mechanisms, resulting in the same functional effect. That proteasome inhibition and IKKbeta inhibition affect distinct molecular pathways downstream of NF-kappaB, both leading to increased chemosensitivity, is previously unreported. Thus, it is conceivable that switching the two principles of NF-kappaB inhibition, once resistance to one of the agents occurs, will improve future treatment regimens.
Collapse
Affiliation(s)
- Katharina Amschler
- Department of Dermatology and Venereology, Georg August University, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Pfaller T, Puntes V, Casals E, Duschl A, Oostingh GJ. In vitro investigation of immunomodulatory effects caused by engineered inorganic nanoparticles – the impact of experimental design and cell choice. Nanotoxicology 2009. [DOI: 10.1080/17435390802546071] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Tobias Pfaller
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Victor Puntes
- Institut Català de Nanotecnologia and Institut Català de Recerca I Estudis Avançats, Barcelona, Spain
| | - Eudald Casals
- Institut Català de Nanotecnologia and Institut Català de Recerca I Estudis Avançats, Barcelona, Spain
| | - Albert Duschl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Gertie J. Oostingh
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| |
Collapse
|
17
|
Schön M, Wienrich BG, Kneitz S, Sennefelder H, Amschler K, Vöhringer V, Weber O, Stiewe T, Ziegelbauer K, Schön MP. KINK-1, a Novel Small-Molecule Inhibitor of IKKβ, and the Susceptibility of Melanoma Cells to Antitumoral Treatment. ACTA ACUST UNITED AC 2008; 100:862-75. [DOI: 10.1093/jnci/djn174] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
18
|
van Beijnum JR, van der Linden E, Griffioen AW. Angiogenic profiling and comparison of immortalized endothelial cells for functional genomics. Exp Cell Res 2008; 314:264-72. [PMID: 17880939 DOI: 10.1016/j.yexcr.2007.08.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 07/30/2007] [Accepted: 08/17/2007] [Indexed: 01/13/2023]
Abstract
Genomics efforts of the past decade have resulted in the identification of numerous genes with putative roles in disease processes, including tumor angiogenesis. To functionally validate these genes, cultured endothelial cells are indispensable tools, though these may not completely mimic the phenotype of tissue endothelial cells as the proper microenvironment is lacking. To obtain experimental data representative of normal physiology, the use of primary endothelial cells is preferred. However, these cells are usually limited in passage number, can be difficult to obtain and show great interindividual variety. Furthermore, transfection efficiency is very limited in primary cells, hampering applications in functional genomics and gene function analysis. The use of properly characterized alternative endothelial cell sources is therefore warranted. Here, we compared immortalized endothelial cells - HMEC, RF24 and EVLC2 - with primary HUVEC. We show that RF24, and to a slightly lesser extent HMEC, resembles primary HUVEC most on all facets examined. RF24, in contrast to EVLC2, express the endothelial markers CD31, CD34, CD105, vWF and VE-cadherin, and are capable of migration and tube formation in vitro. Furthermore, the expression levels of angiogenic growth factors and their receptors are comparable to that of primary EC. In addition, whereas primary HUVEC are resistant to transfection using common lipophilic transfection reagents, HMEC and RF24 could be readily transfected. Hence, these cells pose a valuable tool for functional genomics in angiogenesis research.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Angiogenesis Laboratory, Research Institute for Growth and Development (GROW), Department of Pathology, Maastricht University, 6202AZ, The Netherlands
| | | | | |
Collapse
|