1
|
Liu W, Chen J, Wang T, Gong Y, Yang C, Li Y, Yan X, Duan H, Wang X, Zhang M. Involvement of CD146 in the Cryptococcus neoformans adhesion and infection of brain endothelial cells. Infect Immun 2025; 93:e0014525. [PMID: 40208055 PMCID: PMC12070736 DOI: 10.1128/iai.00145-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Cryptococcal meningitis is a common and refractory central nervous system (CNS) infection with high mortality and disability. For Cryptococcus neoformans (C. neoformans) to penetrate the CNS, it first adheres to and breaches the blood‒brain barrier (BBB). Here, we explored the roles of CD146, an adhesion molecule expressed on the surface of brain microvascular endothelial cells (BMECs), in cryptococcal vascular adhesion and BBB invasion. Following cryptococcal infection, we observed a reduction in CD146 expression in BMECs, which was at least partially mediated by metalloproteinase-9. Once overexpressed on BMECs, CD146 increased C. neoformans adhesion; in contrast, CD146 knockout decreased the attachment of fungi to endothelial cells in vitro. Unexpectedly, CD146 knockout failed to reduce fungal infection in the brain following intravascular instillation of C. neoformans. However, the anti-CD146 antibody AA98 significantly increased the fungemia (spleen CFU), suggesting that CD146 may be involved in the early adhesion and invasion of Cryptococcus into cerebral vessels. AA98, however, failed to extend the survival of C. neoformans infected mice. These results suggest that CD146 may play dispensable roles in the C. neoformans brain infection.
Collapse
Affiliation(s)
- Wei Liu
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junhong Chen
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ting Wang
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Gong
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Yang
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuwei Li
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hongxia Duan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoming Wang
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingshun Zhang
- Department of Immunology, National Vaccine Innovation Platform, NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Urbanczyk M, Abuhelou A, Köninger M, Jeyagaran A, Carvajal-Berrio D, Kim E, Marzi J, Loskill P, Layland SL, Schenke-Layland K. Heterogeneity of Endothelial Cells Impacts the Functionality of Human Pancreatic In Vitro Models. Tissue Eng Part A 2024. [PMID: 39453887 DOI: 10.1089/ten.tea.2024.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024] Open
Abstract
Endothelial cells (ECs) play a crucial role in maintaining tissue homeostasis and functionality. Depending on their tissue of origin, ECs can be highly heterogeneous regarding their morphology, gene and protein expression, functionality, and signaling pathways. Understanding the interaction between organ-specific ECs and their surrounding tissue is therefore critical when investigating tissue homeostasis, disease development, and progression. In vitro models often lack organ-specific ECs, potentially limiting the translatability and validity of the obtained results. The goal of this study was to assess the differences between commonly used EC sources in tissue engineering applications, including human umbilical vein ECs (HUVECs), human dermal microvascular ECs (hdmvECs), and human foreskin microvascular ECs (hfmvECs), and organ-specific human pancreatic microvascular ECs (hpmvECs), and test their impact on functionality within an in vitro pancreas test system used for diabetes research. Utilizing high-resolution Raman microspectroscopy and Raman imaging in combination with established protein and gene expression analyses and exposure to defined physical signals within microfluidic cultures, we identified that ECs exhibit significant differences in their biochemical composition, relevant protein expression, angiogenic potential, and response to the application of mechanical shear stress. Proof-of-concept results showed that the coculture of isolated human islets of Langerhans with hpmvECs significantly increased the functionality when compared with control islets and islets cocultured with HUVECs. Our study demonstrates that the choice of EC type significantly impacts the experimental results, which needs to be considered when implementing ECs into in vitro models.
Collapse
Affiliation(s)
- Max Urbanczyk
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Athar Abuhelou
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marie Köninger
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Abiramy Jeyagaran
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel Carvajal-Berrio
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ellie Kim
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Julia Marzi
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany, Reutlingen, Germany
| | - Peter Loskill
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany, Reutlingen, Germany
- Institute of Biomedical Engineering, Department for Microphysiological Systems, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R Center Tübingen for In Vitro Models and Alternatives to Animal Testing, Tübingen, Germany
| | - Shannon L Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Women's Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany, Reutlingen, Germany
| |
Collapse
|
3
|
Weiß MS, Trapani G, Long H, Trappmann B. Matrix Resistance Toward Proteolytic Cleavage Controls Contractility-Dependent Migration Modes During Angiogenic Sprouting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305947. [PMID: 38477409 PMCID: PMC11109655 DOI: 10.1002/advs.202305947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Tissue homeostasis and disease states rely on the formation of new blood vessels through angiogenic sprouting, which is tightly regulated by the properties of the surrounding extracellular matrix. While physical cues, such as matrix stiffness or degradability, have evolved as major regulators of cell function in tissue microenvironments, it remains unknown whether and how physical cues regulate endothelial cell migration during angiogenesis. To investigate this, a biomimetic model of angiogenic sprouting inside a tunable synthetic hydrogel is created. It is shown that endothelial cells sense the resistance of the surrounding matrix toward proteolytic cleavage and respond by adjusting their migration phenotype. The resistance cells encounter is impacted by the number of covalent matrix crosslinks, crosslink degradability, and the proteolytic activity of cells. When matrix resistance is high, cells switch from a collective to an actomyosin contractility-dependent single cellular migration mode. This switch in collectivity is accompanied by a major reorganization of the actin cytoskeleton, where stress fibers are no longer visible, and F-actin aggregates in large punctate clusters. Matrix resistance is identified as a previously unknown regulator of angiogenic sprouting and, thus, provides a mechanism by which the physical properties of the matrix impact cell migration modes through cytoskeletal remodeling.
Collapse
Affiliation(s)
- Martin S. Weiß
- Bioactive Materials LaboratoryMax Planck Institute for Molecular BiomedicineRöntgenstraße 2048149MünsterGermany
| | - Giuseppe Trapani
- Bioactive Materials LaboratoryMax Planck Institute for Molecular BiomedicineRöntgenstraße 2048149MünsterGermany
| | - Hongyan Long
- Bioactive Materials LaboratoryMax Planck Institute for Molecular BiomedicineRöntgenstraße 2048149MünsterGermany
| | - Britta Trappmann
- Bioactive Materials LaboratoryMax Planck Institute for Molecular BiomedicineRöntgenstraße 2048149MünsterGermany
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn‐Straße 644227DortmundGermany
| |
Collapse
|
4
|
Zhang D, Qiao L. Intestine‐on‐a‐chip for intestinal disease study and pharmacological research. VIEW 2022. [DOI: 10.1002/viw.20220037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Dongxue Zhang
- Department of Chemistry, Institutes of Biomedical Sciences, and Shanghai Stomatological Hospital Fudan University Shanghai China
| | - Liang Qiao
- Department of Chemistry, Institutes of Biomedical Sciences, and Shanghai Stomatological Hospital Fudan University Shanghai China
| |
Collapse
|
5
|
Freitas-Ribeiro S, Diogo GS, Oliveira C, Martins A, Silva TH, Jarnalo M, Horta R, Reis RL, Pirraco RP. Growth Factor-Free Vascularization of Marine-Origin Collagen Sponges Using Cryopreserved Stromal Vascular Fractions from Human Adipose Tissue. Mar Drugs 2022; 20:md20100623. [PMID: 36286447 PMCID: PMC9604698 DOI: 10.3390/md20100623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The successful integration of transplanted three-dimensional tissue engineering (TE) constructs depends greatly on their rapid vascularization. Therefore, it is essential to address this vascularization issue in the initial design of constructs for perfused tissues. Two of the most important variables in this regard are scaffold composition and cell sourcing. Collagens with marine origins overcome some issues associated with mammal-derived collagen while maintaining their advantages in terms of biocompatibility. Concurrently, the freshly isolated stromal vascular fraction (SVF) of adipose tissue has been proposed as an advantageous cell fraction for vascularization purposes due to its highly angiogenic properties, allowing extrinsic angiogenic growth factor-free vascularization strategies for TE applications. In this study, we aimed at understanding whether marine collagen 3D matrices could support cryopreserved human SVF in maintaining intrinsic angiogenic properties observed for fresh SVF. For this, cryopreserved human SVF was seeded on blue shark collagen sponges and cultured up to 7 days in a basal medium. The secretome profile of several angiogenesis-related factors was studied throughout culture times and correlated with the expression pattern of CD31 and CD146, which showed the formation of a prevascular network. Upon in ovo implantation, increased vessel recruitment was observed in prevascularized sponges when compared with sponges without SVF cells. Immunohistochemistry for CD31 demonstrated the improved integration of prevascularized sponges within chick chorioalantoic membrane (CAM) tissues, while in situ hybridization showed human cells lining blood vessels. These results demonstrate the potential of using cryopreserved SVF combined with marine collagen as a streamlined approach to improve the vascularization of TE constructs.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Gabriela S. Diogo
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Catarina Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Tiago H. Silva
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
- Correspondence:
| |
Collapse
|
6
|
Urbanczyk M, Zbinden A, Schenke-Layland K. Organ-specific endothelial cell heterogenicity and its impact on regenerative medicine and biomedical engineering applications. Adv Drug Deliv Rev 2022; 186:114323. [PMID: 35568103 DOI: 10.1016/j.addr.2022.114323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) are a key cellular component of the vascular system as they form the inner lining of the blood vessels. Recent findings highlight that ECs express extensive phenotypic heterogenicity when following the vascular tree from the major vasculature down to the organ capillaries. However, in vitro models, used for drug development and testing, or to study the role of ECs in health and disease, rarely acknowledge this EC heterogenicity. In this review, we highlight the main differences between different EC types, briefly summarize their different characteristics and focus on the use of ECs in in vitro models. We introduce different approaches on how ECs can be utilized in co-culture test systems in the field of brain, pancreas, and liver research to study the role of the endothelium in health and disease. Finally, we discuss potential improvements to current state-of-the-art in vitro models and future directions.
Collapse
|
7
|
Ju Y, Dai X, Tang Z, Ming Z, Ni N, Zhu D, Zhang J, Ma B, Wang J, Huang R, Zhao S, Pang Y, Gu P. Verteporfin-mediated on/off photoswitching functions synergistically to treat choroidal vascular diseases. Bioact Mater 2022; 14:402-415. [PMID: 35386820 PMCID: PMC8964818 DOI: 10.1016/j.bioactmat.2022.01.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022] Open
Abstract
Choroidal vascular diseases, such as age-related macular degeneration, are the leading cause of vision impairment and are characterized by pathological angiogenesis. Verteporfin-mediated photodynamic therapy is a current strategy that selectively occludes choroidal neovasculature. However, the clinically used large-dose systemic administration increases the risk of systemic adverse events, such as phototoxicity to superficial tissues. In this study, we developed an in situ verteporfin delivery system with a photoswitching synergistic function that disassembles in response to intraocular inflammatory enzymes. Under light-on conditions, verteporfin-mediated photodynamic therapy effectively occurs and this leads to vascular occlusion. Under light-off conditions, non-photoactive verteporfin negatively regulates vascular endothelial growth factor-induced angiogenesis as a yes-associated protein inhibitor. Taken together, our system serves as an intraocular verteporfin reservoir to improve the bioavailability of verteporfin by innovatively exploiting its photochemical and biological functions. This work provides a promising strategy with synergistic antiangiogenic effects for the treatment of choroidal vascular diseases. For the first time, an intraocular verteporfin delivery system with on/off photoswitching synergistic functions is reported. VP-TGMS with light-on effectively leads to occlusion of choroidal pathological neovascularization via photodynamic mechanism. VP-TGMS with light-off significantly suppresses VEGF-induced angiogenesis via YAP signaling inhibition. This study provides a promising strategy for the treatment of choroidal vascular diseases.
Collapse
Affiliation(s)
- Yahan Ju
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Xiaochan Dai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Zhimin Tang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Zunzhen Ming
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China
| | - Ni Ni
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Dongqing Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Jing Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Bo Ma
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Jiajing Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Rui Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Siyu Zhao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
| | - Yan Pang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
- Corresponding author. 639 Zhizaoju Rd, Shanghai, 200011, China.
| | - Ping Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, PR China
- Corresponding author. 639 Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
8
|
Alfonsetti M, Castelli V, d’Angelo M, Benedetti E, Allegretti M, Barboni B, Cimini A. Looking for In Vitro Models for Retinal Diseases. Int J Mol Sci 2021; 22:10334. [PMID: 34638674 PMCID: PMC8508697 DOI: 10.3390/ijms221910334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Retina is a layered structure of the eye, composed of different cellular components working together to produce a complex visual output. Because of its important role in visual function, retinal pathologies commonly represent the main causes of visual injury and blindness in the industrialized world. It is important to develop in vitro models of retinal diseases to use them in first screenings before translating in in vivo experiments and clinics. For this reason, it is important to develop bidimensional (2D) models that are more suitable for drug screening and toxicological studies and tridimensional (3D) models, which can replicate physiological conditions, for investigating pathological mechanisms leading to visual loss. This review provides an overview of the most common retinal diseases, relating to in vivo models, with a specific focus on alternative 2D and 3D in vitro models that can replicate the different cellular and matrix components of retinal layers, as well as injury insults that induce retinal disease and loss of the visual function.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | | | - Barbara Barboni
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
9
|
Abstract
Vascularization is a major hurdle in complex tissue and organ engineering. Tissues greater than 200 μm in diameter cannot rely on simple diffusion to obtain nutrients and remove waste. Therefore, an integrated vascular network is required for clinical translation of engineered tissues. Microvessels have been described as <150 μm in diameter, but clinically they are defined as <1 mm. With new advances in super microsurgery, vessels less than 1 mm can be anastomosed to the recipient circulation. However, this technical advancement still relies on the creation of a stable engineered microcirculation that is amenable to surgical manipulation and is readily perfusable. Microvascular engineering lays on the crossroads of microfabrication, microfluidics, and tissue engineering strategies that utilize various cellular constituents. Early research focused on vascularization by co-culture and cellular interactions, with the addition of angiogenic growth factors to promote vascular growth. Since then, multiple strategies have been utilized taking advantage of innovations in additive manufacturing, biomaterials, and cell biology. However, the anatomy and dynamics of native blood vessels has not been consistently replicated. Inconsistent results can be partially attributed to cell sourcing which remains an enigma for microvascular engineering. Variations of endothelial cells, endothelial progenitor cells, and stem cells have all been used for microvascular network fabrication along with various mural cells. As each source offers advantages and disadvantages, there continues to be a lack of consensus. Furthermore, discord may be attributed to incomplete understanding about cell isolation and characterization without considering the microvascular architecture of the desired tissue/organ.
Collapse
|
10
|
Ringland C, Schweig JE, Paris D, Shackleton B, Lynch CE, Eisenbaum M, Mullan M, Crawford F, Abdullah L, Bachmeier C. Apolipoprotein E isoforms differentially regulate matrix metallopeptidase 9 function in Alzheimer's disease. Neurobiol Aging 2020; 95:56-68. [PMID: 32758917 DOI: 10.1016/j.neurobiolaging.2020.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 01/10/2023]
Abstract
Apolipoprotein E (APOE) has been shown to influence amyloid-β (Aβ) clearance from the brain in an isoform-specific manner. Our prior work showed that Aβ transit across the blood-brain-barrier was reduced by apoE4, compared to other apoE isoforms, due to elevated lipoprotein receptor shedding in brain endothelia. Recently, we demonstrated that matrix metallopeptidase 9 (MMP-9) induces lipoprotein receptor proteolysis in an apoE isoform-dependent manner, which impacts Aβ elimination from the brain. The current studies interrogated the relationship between apoE and MMP-9 and found that apoE impacted proMMP-9 cellular secretion from brain endothelia (apoE2 < apoE3 = apoE4). In a cell-free assay, apoE dose-dependently reduced MMP-9 activity, with apoE4 showing a significantly weaker ability to inhibit MMP-9 function than apoE2 or apoE3. Finally, we observed elevated MMP-9 expression and activity in the cerebrovasculature of both human and animal AD brain specimens with an APOE4 genotype. Collectively, these findings suggest a role for apoE in regulating MMP-9 disposition and may describe the effect of apoE4 on Aβ pathology in the AD brain.
Collapse
Affiliation(s)
- Charis Ringland
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK.
| | | | | | | | | | - Maxwell Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Laila Abdullah
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
11
|
Ocaña MC, Martínez-Poveda B, Quesada AR, Medina MÁ. Highly Glycolytic Immortalized Human Dermal Microvascular Endothelial Cells are Able to Grow in Glucose-Starved Conditions. Biomolecules 2019; 9:biom9080332. [PMID: 31374952 PMCID: PMC6723428 DOI: 10.3390/biom9080332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells form the inner lining of blood vessels, in a process known as angiogenesis. Excessive angiogenesis is a hallmark of several diseases, including cancer. The number of studies in endothelial cell metabolism has increased in recent years, and new metabolic targets for pharmacological treatment of pathological angiogenesis are being proposed. In this work, we wanted to address experimental evidence of substrate (namely glucose, glutamine and palmitate) dependence in immortalized dermal microvascular endothelial cells in comparison to primary endothelial cells. In addition, due to the lack of information about lactate metabolism in this specific type of endothelial cells, we also checked their capability of utilizing extracellular lactate. For fulfilling these aims, proliferation, migration, Seahorse, substrate uptake/utilization, and mRNA/protein expression experiments were performed. Our results show a high glycolytic capacity of immortalized dermal microvascular endothelial cells, but an early independence of glucose for cell growth, whereas a total dependence of glutamine to proliferate was found. Additionally, in contrast with reported data in other endothelial cell lines, these cells lack monocarboxylate transporter 1 for extracellular lactate incorporation. Therefore, our results point to the change of certain metabolic features depending on the endothelial cell line.
Collapse
Affiliation(s)
- Mª Carmen Ocaña
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
| | - Beatriz Martínez-Poveda
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
| | - Ana R Quesada
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Málaga, Spain
| | - Miguel Ángel Medina
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain.
- IBIMA (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain.
- CIBER de Enfermedades Raras (CIBERER), E-29071 Málaga, Spain.
| |
Collapse
|
12
|
Li WD, Zhou DM, Sun LL, Xiao L, Liu Z, Zhou M, Wang WB, Li XQ. LncRNA WTAPP1 Promotes Migration and Angiogenesis of Endothelial Progenitor Cells via MMP1 Through MicroRNA 3120 and Akt/PI3K/Autophagy Pathways. Stem Cells 2018; 36:1863-1874. [PMID: 30171660 DOI: 10.1002/stem.2904] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/22/2018] [Accepted: 07/27/2018] [Indexed: 01/17/2023]
Affiliation(s)
- Wen-Dong Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Dong-Ming Zhou
- Department of Hematology, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Li-Li Sun
- Department of Vascular Surgery; The Second Affiliated Hospital of Soochow University; Suzhou JiangSu People's Republic of China
| | - Lun Xiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Zhao Liu
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Min Zhou
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| | - Wen-Bin Wang
- Department of General Surgery; The Fourth Affiliated Hospital of Anhui Medical University; HeFei People's Republic of China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital; Nanjing University Medical School; Nanjing JiangSu People's Republic of China
| |
Collapse
|
13
|
Pogontke C, Guadix JA, Ruiz-Villalba A, Pérez-Pomares JM. Development of the Myocardial Interstitium. Anat Rec (Hoboken) 2018; 302:58-68. [PMID: 30288955 DOI: 10.1002/ar.23915] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/26/2018] [Accepted: 05/11/2018] [Indexed: 12/24/2022]
Abstract
The space between cardiac myocytes is commonly referred-to as the cardiac interstitium (CI). The CI is a unique, complex and dynamic microenvironment in which multiple cell types, extracellular matrix molecules, and instructive signals interact to crucially support heart homeostasis and promote cardiac responses to normal and pathologic stimuli. Despite the biomedical and clinical relevance of the CI, its detailed cellular structure remains to be elucidated. In this review, we will dissect the organization of the cardiac interstitium by following its changing cellular and molecular composition from embryonic developmental stages to adulthood, providing a systematic analysis of the biological components of the CI. The main goal of this review is to contribute to our understanding of the CI roles in health and disease. Anat Rec, 302:58-68, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cristina Pogontke
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| | - Juan A Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| | - Adrián Ruiz-Villalba
- Stem Cell Therapy Area, Foundation for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - José M Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| |
Collapse
|
14
|
Wenz A, Tjoeng I, Schneider I, Kluger PJ, Borchers K. Improved vasculogenesis and bone matrix formation through coculture of endothelial cells and stem cells in tissue-specific methacryloyl gelatin-based hydrogels. Biotechnol Bioeng 2018; 115:2643-2653. [PMID: 29981277 DOI: 10.1002/bit.26792] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/12/2018] [Accepted: 07/02/2018] [Indexed: 01/21/2023]
Abstract
The coculture of osteogenic and angiogenic cells and the resulting paracrine signaling via soluble factors are supposed to be crucial for successfully engineering vascularized bone tissue equivalents. In this study, a coculture system combining primary human adipose-derived stem cells (hASCs) and primary human dermal microvascular endothelial cells (HDMECs) within two types of hydrogels based on methacryloyl-modified gelatin (GM) as three-dimensional scaffolds was examined for its support of tissue specific cell functions. HDMECs, together with hASCs as supporting cells, were encapsulated in soft GM gels and were indirectly cocultured with hASCs encapsulated in stiffer GM hydrogels additionally containing methacrylate-modified hyaluronic acid and hydroxyapatite particles. After 14 days, the hASC in the stiffer gels (constituting the "bone gels") expressed matrix proteins like collagen type I and fibronectin, as well as bone-specific proteins osteopontin and alkaline phosphatase. After 14 days of coculture with HDMEC-laden hydrogels, the viscoelastic properties of the bone gels were significantly higher compared with the gels in monoculture. Within the soft vascularization gels, the formed capillary-like networks were significantly longer after 14 days of coculture than the structures in the control gels. In addition, the stability as well as the complexity of the vascular networks was significantly increased by coculture. We discussed and concluded that osteogenic and angiogenic signals from the culture media as well as from cocultured cell types, and tissue-specific hydrogel composition all contribute to stimulate the interplay between osteogenesis and angiogenesis in vitro and are a basis for engineering vascularized bone.
Collapse
Affiliation(s)
- Annika Wenz
- Department of Materials Science, Institute of Interfacial Engineering and Plasmatechnology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Iva Tjoeng
- Department of Interfacial Engineering and Material Science, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Isabelle Schneider
- Department of Interfacial Engineering and Material Science, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Petra J Kluger
- Department of Interfacial Engineering and Material Science, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany.,Department of Smart Biomaterials, Reutlingen University, Reutlingen, Germany
| | - Kirsten Borchers
- Department of Materials Science, Institute of Interfacial Engineering and Plasmatechnology IGVP, University of Stuttgart, Stuttgart, Germany.,Department of Interfacial Engineering and Material Science, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| |
Collapse
|
15
|
Murugavel S, Bugyei-Twum A, Matkar PN, Al-Mubarak H, Chen HH, Adam M, Jain S, Narang T, Abdin RM, Qadura M, Connelly KA, Leong-Poi H, Singh KK. Valproic Acid Induces Endothelial-to-Mesenchymal Transition-Like Phenotypic Switching. Front Pharmacol 2018; 9:737. [PMID: 30050438 PMCID: PMC6050396 DOI: 10.3389/fphar.2018.00737] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, is a widely used anticonvulsant drug that is currently undergoing clinical evaluation for anticancer therapy due to its anti-angiogenic potential. Endothelial cells (ECs) can transition into mesenchymal cells and this form of EC plasticity is called endothelial-to-mesenchymal transition (EndMT), which is widely implicated in several pathologies including cancer and organ fibrosis. However, the effect of VPA on EC plasticity and EndMT remains completely unknown. We report herein that VPA-treatment significantly inhibits tube formation, migration, nitric oxide production, proliferation and migration in ECs. A microscopic evaluation revealed, and qPCR, immunofluorescence and immunoblotting data confirmed EndMT-like phenotypic switching as well as an increased expression of pro-fibrotic genes in VPA-treated ECs. Furthermore, our data confirmed important and regulatory role played by TGFβ-signaling in VPA-induced EndMT. Our qPCR array data performed for 84 endothelial genes further supported our findings and demonstrated 28 significantly and differentially regulated genes mainly implicated in angiogenesis, endothelial function, EndMT and fibrosis. We, for the first time report that VPA-treatment associated EndMT contributes to the VPA-associated loss of endothelial function. Our data also suggest that VPA based therapeutics may exacerbate endothelial dysfunction and EndMT-related phenotype in patients undergoing anticonvulsant or anticancer therapy, warranting further investigation.
Collapse
Affiliation(s)
| | - Antoinette Bugyei-Twum
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Pratiek N Matkar
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Husain Al-Mubarak
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Hao H Chen
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mohamed Adam
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Shubha Jain
- Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Tanya Narang
- Faculty of Science, York University, Toronto, ON, Canada
| | - Rawand M Abdin
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Mohammad Qadura
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Kim A Connelly
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Howard Leong-Poi
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Krishna K Singh
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
16
|
Amtul Z, Yang J, Nikolova S, Lee TY, Bartha R, Cechetto DF. The Dynamics of Impaired Blood-Brain Barrier Restoration in a Rat Model of Co-morbid Injury. Mol Neurobiol 2018; 55:8071-8083. [PMID: 29508280 DOI: 10.1007/s12035-018-0904-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Defect in brain microperfusion is increasingly recognized as an antecedent event to Alzheimer's disease (AD) and ischemia. Nevertheless, studies on the role of impaired microperfusion as a pathological trigger to neuroinflammation, Aβ deposition as well as blood-brain barrier (BBB) disruption, and the etiological link between AD and ischemia are lacking. In this study, we employ in vivo sequential magnetic resonance imaging (MRI) and computed tomography (CT) imaging in a co-morbid rat model of β-amyloid toxicity (Aβ) and ischemia (ET1) with subsequent histopathology of striatal lesion core and penumbra at 1, 7, and 28 days post injury. Within 24 h, cerebral injury resulted in increased BBB permeability due to the dissolution of β-dystroglycan (β-DG) and basement membrane laminin by active matrix metalloproteinase9 (MMP9). As a result, net flow of circulating IgG down a hydrostatic gradient into the parenchyma led to vasogenic edema and impaired perfusion, thus increasing the apparent hyperintensity in true fast imaging with steady-state free precession (true FISP) imaging and acute hypoperfusion in CT. This was followed by a slow recruitment of reactive astroglia to the affected brain and depolarization of aquaporin4 (AQP4) expression resulting in cytotoxic edema-in an attempt to resolve vasogenic edema. On d28, functional BBB was restored in ET1 rats as observed by astrocytic MMP9 release, β-DG stabilization, and new vessel formation. This was confirmed by reduced hyperintensity on true FISP imaging and normalized cerebral blood flow in CT. While, Aβ toxicity alone was not detrimental enough, Aβ+ET1 rats showed delayed differential expression of MMP9, late recruitment of astroglial cells, protracted loss of AQP4 depolarization, and thus delayed BBB restoration and cerebral perfusion.
Collapse
Affiliation(s)
- Zareen Amtul
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 5C1, Canada.
| | - Jun Yang
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Simona Nikolova
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Ting-Yim Lee
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Robert Bartha
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada.,Department of Medical Biophysics, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - David F Cechetto
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 5C1, Canada
| |
Collapse
|
17
|
Iwata Y, Klaren WD, Lebakken CS, Grimm FA, Rusyn I. High-Content Assay Multiplexing for Vascular Toxicity Screening in Induced Pluripotent Stem Cell-Derived Endothelial Cells and Human Umbilical Vein Endothelial Cells. Assay Drug Dev Technol 2017; 15:267-279. [PMID: 28771372 DOI: 10.1089/adt.2017.786] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells (ECs) play a major role in blood vessel formation and function. While there is longstanding evidence for the potential of chemical exposures to adversely affect EC function and vascular development, the hazard potential of chemicals with respect to vascular effects is not routinely evaluated in safety assessments. Induced pluripotent stem cell (iPSC)-derived ECs promise to provide a physiologically relevant, organotypic culture model that is amenable for high-throughput (HT) EC toxicant screening and may represent a viable alternative to traditional in vitro models, including human umbilical vein endothelial cells (HUVECs). To evaluate the utility of iPSC-ECs for multidimensional HT toxicity profiling of chemicals, both iPSC-ECs and HUVECs were exposed to selected positive (angiogenesis inhibitors, cytotoxic agents) and negative compounds in concentration response for either 16 or 24 h in a 384-well plate format. Furthermore, chemical effects on vascularization were quantified using EC angiogenesis on biological (Geltrex™) and synthetic (SP-105 angiogenesis hydrogel) extracellular matrices. Cellular toxicity was assessed using high-content live cell imaging and the CellTiter-Glo® assay. Assay performance indicated good to excellent assay sensitivity and reproducibility for both cell types investigated. Both iPSC-derived ECs and HUVECs formed tube-like structures on Geltrex™ and hydrogel, an effect that was inhibited by angiogenesis inhibitors and cytotoxic agents in a concentration-dependent manner. The quality of HT assays in HUVECs was generally higher than that in iPSC-ECs. Altogether, this study demonstrates the capability of ECs for comprehensive assessment of the biological effects of chemicals on vasculature in a HT compatible format.
Collapse
Affiliation(s)
- Yasuhiro Iwata
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - William D Klaren
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | | | - Fabian A Grimm
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Ivan Rusyn
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| |
Collapse
|
18
|
Inspiration from heart development: Biomimetic development of functional human cardiac organoids. Biomaterials 2017; 142:112-123. [PMID: 28732246 DOI: 10.1016/j.biomaterials.2017.07.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/02/2023]
Abstract
Recent progress in human organoids has provided 3D tissue systems to model human development, diseases, as well as develop cell delivery systems for regenerative therapies. While direct differentiation of human embryoid bodies holds great promise for cardiac organoid production, intramyocardial cell organization during heart development provides biological foundation to fabricate human cardiac organoids with defined cell types. Inspired by the intramyocardial organization events in coronary vasculogenesis, where a diverse, yet defined, mixture of cardiac cell types self-organizes into functional myocardium in the absence of blood flow, we have developed a defined method to produce scaffold-free human cardiac organoids that structurally and functionally resembled the lumenized vascular network in the developing myocardium, supported hiPSC-CM development and possessed fundamental cardiac tissue-level functions. In particular, this development-driven strategy offers a robust, tunable system to examine the contributions of individual cell types, matrix materials and additional factors for developmental insight, biomimetic matrix composition to advance biomaterial design, tissue/organ-level drug screening, and cell therapy for heart repair.
Collapse
|
19
|
Effect of Cytokines on the Formation Tube-Like Structures by Endothelial Cells in the Presence of Trophoblast Cells. Bull Exp Biol Med 2017; 163:148-158. [PMID: 28577098 DOI: 10.1007/s10517-017-3756-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Indexed: 12/16/2022]
Abstract
Despite ample data on cytokine secretion in the uteroplacental interface, the influence of microenvironment cells, in particular, trophoblast cells on angiogenesis and the role of cytokines in this process remain poorly studied. We studied the influence of cytokines on the formation of tube-like structures by endothelial cells in the presence of trophoblast cells and showed that trophoblast cells suppressed the angiogenic potential of endothelial cells. Antiangiogenic cytokines IFN-γ, IL-10, TNF-α, and TGFβ via modulation of trophoblast cells stimulated the formation of tube-like structures by endothelial cells. In the co-culture of endothelial and trophoblast cells, the effects of cytokines changed and they gained additional regulatory functions.
Collapse
|
20
|
Gunatillake T, Yong HEJ, Dunk CE, Keogh RJ, Borg AJ, Cartwright JE, Whitley GS, Murthi P. Homeobox gene TGIF-1 is increased in placental endothelial cells of human fetal growth restriction. Reproduction 2016; 152:457-65. [PMID: 27539603 DOI: 10.1530/rep-16-0068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/18/2016] [Indexed: 01/09/2023]
Abstract
Aberrant placental angiogenesis is associated with fetal growth restriction (FGR). In mice, targeted disruption of the homeobox gene, transforming growth β-induced factor (Tgif-1), which is also a transcription factor, causes defective placental vascularisation. Nevertheless, the role of TGIF-1 in human placental angiogenesis is unclear. We have previously reported increased TGIF-1 expression in human FGR placentae and demonstrated localisation of TGIF-1 protein in placental endothelial cells (ECs). However, its functional role remains to be investigated. In this study, we aimed to specifically compare TGIF-1 mRNA expression in placental ECs isolated from human FGR-affected pregnancies with gestation-matched control pregnancies in two independent cohorts from Australia and Canada and to identify the functional role of TGIF-1 in placental angiogenesis using the human umbilical vein endothelial cell-derived cell line, SGHEC-7, and primary human umbilical vein ECs. Real-time PCR revealed that TGIF-1 mRNA expression was significantly increased in ECs isolated from FGR-affected placentae compared with that of controls. The functional roles of TGIF-1 were determined in ECs after TGIF-1 siRNA transfection. TGIF-1 inactivation in ECs significantly reduced TGIF-1 at both the mRNA and protein levels, as well as the proliferative and invasive potential, but significantly increased the angiogenic potential. Using angiogenesis PCR screening arrays, we identified ITGAV, NRP-1, ANPGT-1 and ANPGT-2 as novel downstream targets of TGIF-1, after TGIF-1 inactivation in ECs. Collectively, these results show that TGIF-1 regulates EC function and the expression of angiogenic molecules; and when abnormally expressed, may contribute to the aberrant placental angiogenesis observed in FGR.
Collapse
Affiliation(s)
- Tilini Gunatillake
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Hannah E J Yong
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Caroline E Dunk
- Lunenfeld Tanenbaum-Research InstituteMount Sinai Hospital, Toronto, Ontario, Canada
| | - Rosemary J Keogh
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Borg
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia
| | - Judith E Cartwright
- Institute of Cardiovascular and Cell SciencesSt George's, University of London, London, UK
| | - Guy S Whitley
- Institute of Cardiovascular and Cell SciencesSt George's, University of London, London, UK
| | - Padma Murthi
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia Department of MedicineSchool of Clinical Sciences, Monash University, Clayton, Victoria, Australia The Ritchie CentreHudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
21
|
Sanz-Nogués C, O'Brien T. In vitro models for assessing therapeutic angiogenesis. Drug Discov Today 2016; 21:1495-1503. [PMID: 27262402 DOI: 10.1016/j.drudis.2016.05.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/30/2016] [Accepted: 05/25/2016] [Indexed: 01/05/2023]
Abstract
Arterial obstruction leading to ischemia causes a reduction of oxygen and nutrient supply to distal tissues. The physiological response to tissue ischemia triggers a cascade of events that results in the development of accessory vasculature to increase local tissue perfusion and to salvage tissue. However, this adaptive mechanism of repair is suboptimal in some patients. Therapeutic angiogenesis aims to stimulate new blood vessel formation via the local administration of proangiogenic agents or cell therapy products (CTPs). In this review, we provide a summary of the current understanding of in vitro models for assessing the angiogenic potential of a product.
Collapse
Affiliation(s)
- Clara Sanz-Nogués
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, National University of Ireland Galway, Newcastle Road, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, National University of Ireland Galway, Newcastle Road, Galway, Ireland.
| |
Collapse
|
22
|
Kang Y, Mochizuki N, Khademhosseini A, Fukuda J, Yang Y. Engineering a vascularized collagen-β-tricalcium phosphate graft using an electrochemical approach. Acta Biomater 2015; 11:449-58. [PMID: 25263031 DOI: 10.1016/j.actbio.2014.09.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 09/16/2014] [Accepted: 09/21/2014] [Indexed: 12/30/2022]
Abstract
Vascularization of three-dimensional large synthetic grafts for tissue regeneration remains a significant challenge. Here we demonstrate an electrochemical approach, named the cell electrochemical detachment (CED) technique, to form an integral endothelium and use it to prevascularize a collagen-β-tricalcium phosphate (β-TCP) graft. The CED technique electrochemically detached an integral endothelium from a gold-coated glass rod to a collagen-infiltrated, channeled, macroporous β-TCP scaffold, forming an endothelium-lined microchannel containing graft upon removal of the rod. The in vitro results from static and perfusion culture showed that the endothelium robustly emanated microvascular sprouting and prevascularized the entire collagen/β-TCP integrated graft. The in vivo subcutaneous implantation studies showed that the prevascularized collagen/β-TCP grafts established blood flow originating from the endothelium-lined microchannel within a week, and the blood flow covered more areas in the graft over time. In addition, many blood vessels invaded the prevascularized collagen/β-TCP graft and the in vitro preformed microvascular networks anastomosed with the host vasculature, while collagen alone without the support of rigid ceramic scaffold showed less blood vessel invasion and anastomosis. These results suggest a promising strategy for effectively vascularizing large tissue-engineered grafts by integrating multiple hydrogel-based CED-engineered endothelium-lined microchannels into a rigid channeled macroporous scaffold.
Collapse
|
23
|
The influence of simulated microgravity on purinergic signaling is different between individual culture and endothelial and smooth muscle cell coculture. BIOMED RESEARCH INTERNATIONAL 2014; 2014:413708. [PMID: 25243140 PMCID: PMC4163442 DOI: 10.1155/2014/413708] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/30/2014] [Accepted: 07/23/2014] [Indexed: 01/10/2023]
Abstract
Exposure to microgravity conditions causes cardiovascular deconditioning in astronauts during spaceflight. Until now, no specific drugs are available for countermeasure, since the underlying mechanism is largely unknown. Endothelial cells (ECs) and smooth muscle cells (SMCs) play key roles in various vascular functions, many of which are regulated by purinergic 2 (P2) receptors. However, their function in ECs and SMCs under microgravity conditions is still unclear. In this study, primary ECs and SMCs were isolated from bovine aorta and verified with specific markers. We show for the first time that the P2 receptor expression pattern is altered in ECs and SMCs after 24 h exposure to simulated microgravity using a clinostat. However, conditioned medium compensates this change in specific P2 receptors, for example, P2X7. Notably, P2 receptors such as P2X7 might be the important players during the paracrine interaction. Additionally, ECs and SMCs secreted different cytokines under simulated microgravity, leading into a pathogenic proliferation and migration. In conclusion, our data indicate P2 receptors might be important players responding to gravity changes in ECs and SMCs. Since some artificial P2 receptor ligands are applied as drugs, it is reasonable to assume that they might be promising candidates against cardiovascular deconditioning in the future.
Collapse
|
24
|
Mansouri K, Khodarahmi R, Ghadami SA. An in vitro model for spontaneous angiogenesis using rat mesenteric endothelial cells: possible therapeutic perspective for obesity and related disorders. PHARMACEUTICAL BIOLOGY 2013; 51:974-980. [PMID: 23735119 DOI: 10.3109/13880209.2013.773452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
CONTEXT Abnormal obesity and the related diseases, such as diabetes and cardiovascular disease, are the main causes of mortality, around the world. A key feature of the adipogenesis and obesity is angiogenesis-dependent tissue growth accompanied with extracellular remodeling. In this way, suppression of angiogenesis may be a key point for preventing the adipogenesis. OBJECTIVE In the present study, to provide a deeper insight to understand obesity and screening for more effective therapeutics, we have developed a three-dimensional in vitro model of microvessel formation under collagen matrix culture using endothelial cells, extracted from a suitable tissue. MATERIALS AND METHODS In a successful approach for developing an angiogenesis model, the rat mesenteric microvascular endothelial cells (RMMECs) were isolated, coated on dextran beads and then suspended in collagen gel. Additionally, the proliferation as well as migration of endothelial cells were analyzed and compared with human umbilical vein endothelial cells (HUVECs). RESULTS RMMECs showed remarkable migration ability and had higher growth during the logarithmic growth phase, when compared with HUVECs. Also, no significance differences in morphogenesis were observed between HUVECs and RMMECs. DISCUSSION AND CONCLUSION The model may be useful in providing insights to develop potential intervention strategies in vivo against obesity-related disorders. Targeting endothelial cells is an interesting and exciting possibility that may be raised in further investigations.
Collapse
Affiliation(s)
- Kamran Mansouri
- Molecular Medicine Department, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
25
|
Abstract
In most humans, obesity is associated with a chronic low-grade inflammatory reaction occurring in several organ tissues, including the adipose tissue. Infiltration of bone marrow derived leukocytes (granulocytes, monocytes, lymphocytes) into expanding adipose depots appears to be an integral component of inflammation in obesity. Circulating leukocytes invade organ tissues mainly through post-capillary venules in the microcirculation. The endothelium of the post-capillary venules acts as a gatekeeper to leukocyte adhesion and extravasation by displacing on its luminal surface adhesion molecules that bind the adhesive receptors expressed on circulating leukocytes. Several studies investigating the impact of obesity on the microcirculation have demonstrated the occurrence of microvascular dysfunction in experimental animal model of obesity, as well as in obese humans. To date though, working hypotheses and study designs have favored the view that microvascular alterations are secondary to adipose tissue dysfunction. Indeed, a significant amount of data exists in the scientific literature to support the concept that microvascular dysfunction may precede and cause adipose tissue inflammation in obesity. Through review of key published data, this article prospectively presents the concept that in response to nutrients overload the vascular endothelium of the microcirculation acutely activates inflammatory pathways that initiate infiltration of leukocytes in visceral adipose tissue, well before weight gain and overt obesity. The anatomical and physiological heterogeneity of different microcirculations is also discussed toward the understanding of how obesity induces different inflammatory phenotypes in visceral and subcutaneous fat depots.
Collapse
Affiliation(s)
- Rosario Scalia
- Department of Physiology and Cardiovascular Research Center, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
26
|
Retinol-binding protein 4 induces inflammation in human endothelial cells by an NADPH oxidase- and nuclear factor kappa B-dependent and retinol-independent mechanism. Mol Cell Biol 2012; 32:5103-15. [PMID: 23071093 DOI: 10.1128/mcb.00820-12] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Serum retinol-binding protein 4 (RBP4) is the sole specific vitamin A (retinol) transporter in blood. Elevation of serum RBP4 in patients has been linked to cardiovascular disease and diabetic retinopathy. However, the significance of RBP4 elevation in the pathogenesis of these vascular diseases is unknown. Here we show that RBP4 induces inflammation in primary human retinal capillary endothelial cells (HRCEC) and human umbilical vein endothelial cells (HUVEC) by stimulating expression of proinflammatory molecules involved in leukocyte recruitment and adherence to endothelium, including vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), E-selectin, monocyte chemoattractant protein 1 (MCP-1), and interleukin-6 (IL-6). We demonstrate that these novel effects of RBP4 are independent of retinol and the RBP4 membrane receptor STRA6 and occur in part via activation of NADPH oxidase and NF-κB. Importantly, retinol-free RBP4 (apo-RBP4) was as potent as retinol-bound RBP4 (holo-RBP4) in inducing proinflammatory molecules in both HRCEC and HUVEC. These studies reveal that RBP4 elevation can directly contribute to endothelial inflammation and therefore may play a causative role in the development or progression of vascular inflammation during cardiovascular disease and microvascular complications of diabetes.
Collapse
|
27
|
Lu Z, Li Y, Jin J, Zhang X, Lopes-Virella MF, Huang Y. Toll-like receptor 4 activation in microvascular endothelial cells triggers a robust inflammatory response and cross talk with mononuclear cells via interleukin-6. Arterioscler Thromb Vasc Biol 2012; 32:1696-706. [PMID: 22596222 DOI: 10.1161/atvbaha.112.251181] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE It is known that toll-like receptor 4 (TLR4) plays an important role in atherosclerosis. Because both microvascular (MIC) and macrovascular (MAC) endothelial cells (ECs) are present in atherosclerotic lesions, the present study compared TLR4-triggered inflammatory response and cross talk with mononuclear cells between MIC and MAC ECs. METHODS AND RESULTS ELISA, real-time polymerase chain reaction, and gene expression profiling showed that TLR4 activation by lipopolysaccharide stimulated a much higher expression of inflammatory genes including cytokines, chemokines, growth factors, and adhesion molecules in MIC ECs than MAC ECs. Furthermore, coculture studies showed that TLR4 activation in MIC ECs, but not MAC ECs, induced a cross talk with U937 mononuclear cells through MIC EC-released interleukin-6 to upregulate matrix metalloproteinase-1 expression in U937 cells. To explore molecular mechanisms underlying the different responses to TLR4 activation between MIC and MAC ECs, we showed that MIC ECs had a higher expression of TLR4 and CD14 and a higher TLR4-mediated nuclear factor-kappaB activity than MAC ECs. CONCLUSIONS The present study showed that TLR4 activation triggers a more robust inflammatory response in MIC ECs than MAC ECs. Given the importance of inflammatory cytokines and matrix metalloproteinases in plaque rupture, MIC ECs may play a key role in plaque destabilization through a TLR4-dependent mechanism.
Collapse
Affiliation(s)
- Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | | | | | | | | | | |
Collapse
|
28
|
Kozlosky J, Bonventre J, Cooper K. Methyl tert butyl ether is anti-angiogenic in both in vitro and in vivo mammalian model systems. J Appl Toxicol 2012; 33:820-7. [PMID: 22407988 DOI: 10.1002/jat.2737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 01/05/2012] [Accepted: 01/14/2012] [Indexed: 11/09/2022]
Abstract
Methyl-tertiary butyl ether (MTBE), a well known gasoline oxygenate, and US Food and Drug Administration approved gallstone treatment, has been previously shown to specifically target teleost embryonic angiogenesis. The studies reported here were to determine whether similar vascular disrupting effects occur in higher vertebrate models. Rat brain endothelial cells were isolated and allowed to form microcapillary-like tubes on Matrigel. MTBE (0.34-34.0 mm) exposure resulted in a dose-dependent reduction of tube formation, with the LOAEL at 0.34 mm, while MTBE's primary metabolite, tertiary butyl alcohol had no effect on tube formation. HUVECs, a primary cell line representing macrovascular cells, were able to form tubes on Matrigel in the presence of MTBE (1.25-80 mm), but the tubes were narrower than those formed in the absence of MTBE. In a mouse Matrigel plug implantation assay, 34.0 mm MTBE completely inhibited vessel invasion into plugs containing endothelial cell growth supplement (ECGS) compared with control plugs with ECGS alone. When timed-pregnant Fisher 344 rats were gavaged with MTBE (500-1500 mg kg(-1) ) from day 6 of organogenesis through 10 days post-parturition, no organ toxicity or histological changes in pup vasculature were observed. Results of the in vitro cell culture studies show that MTBE is anti-angiogenic at mm concentrations and has potential use as an anti-angiogenic treatment for solid tumors with minimal toxicity.
Collapse
Affiliation(s)
- John Kozlosky
- Environmental Science, Rutgers University, New Brunswick, NJ, USA
| | | | | |
Collapse
|
29
|
Yoshida D, Noha M, Watanabe K, Sugisaki Y, Teramoto A. Novel approach to analysis of in vitro tumor angiogenesis with a variable-pressure scanning electron microscope: suppression by matrix metalloproteinase inhibitor SI-27. Brain Tumor Pathol 2012; 18:89-100. [PMID: 11908879 DOI: 10.1007/bf02479421] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Degradation of basement membrane by metalloproteinases (MMP) is a critical step in tumor angiogenesis. To evaluate in vitro angiogenesis, several models have been employed, including bovine cornea, fenestrated rat brain, Matrigel, and others. These models did not provide quantitative analysis of capillary formation. The current study aimed for a novel approach to in vitro assay of angiogenesis with a "wet scanning electron microscope (SEM)" to investigate suppression of tumor angiogenesis by the MMP inhibitor, SI-27. The effects of noncytotoxic concentrations of SI-27 (1-100 microM) were determined on nonmitogenic vascular endothelial growth factor (VEGF) (10 ng/ml)-mediated cell motility and in vitro angiogenesis of human umbilical vein endothelial cells (HUVECs). Activities of MMP and tissue inhibitor of metalloproteinase (TIMP) were determined by enzyme-linked immunosorbent assay (ELISA). Subsequently, the inhibitory effect of SI-27 was examined on in vitro angiogenesis stimulated by supernatants of human glioma cell lines (U87MG, U251MG, or U373MG). In vitro angiogenesis was quantitatively analyzed with a variable-pressure SEM. Cell motility and in vitro angiogenesis by HUVECs were significantly increased by VEGF along with elevated MMP-1 and -2 activity, whereas SI-27 significantly suppressed VEGF-mediated in vitro angiogenesis and inactivated both MMP-1 and MMP-2, but not inhibited cell motility. The angiogenesis promoted by glioma supernatants showed a significant reduction in the presence of SI-27. SI-27, a novel MMP inhibitor, inhibited tumor angiogenesis in vitro. It can be anticipated to prevent tumor growth through its angiosuppressive effect. Quantitative analysis with a variable-pressure SEM is a novel approach to in vitro angiogenesis assay.
Collapse
Affiliation(s)
- D Yoshida
- Department of Neurosurgery, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
30
|
Duran-Vilaregut J, del Valle J, Manich G, Camins A, Pallàs M, Vilaplana J, Pelegrí C. Role of matrix metalloproteinase-9 (MMP-9) in striatal blood-brain barrier disruption in a 3-nitropropionic acid model of Huntington's disease. Neuropathol Appl Neurobiol 2011; 37:525-37. [DOI: 10.1111/j.1365-2990.2010.01157.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
31
|
Freidja ML, Toutain B, Caillon A, Desquiret V, Lambert D, Loufrani L, Procaccio V, Henrion D. Heme oxygenase 1 is differentially involved in blood flow-dependent arterial remodeling: role of inflammation, oxidative stress, and nitric oxide. Hypertension 2011; 58:225-31. [PMID: 21690482 DOI: 10.1161/hypertensionaha.111.170266] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heme oxygenase 1 is induced by hemodynamic forces in vascular smooth muscle and endothelial cells. We investigated the involvement of heme oxygenase 1 in flow (shear stress)-dependent remodeling. Two or 14 days after ligation of mesenteric resistance arteries, vessels were isolated. In rats, at 14 days, diameter increased by 23% in high-flow arteries and decreased by 22% in low-flow arteries compared with normal flow vessels. Heme oxygenase activity inhibition using Tin-protoporphyrin abolished diameter enlargement in high-flow arteries and accentuated arterial narrowing in low-flow arteries (32% diameter decrease versus 22% in control). Two days after ligation, heme oxygenase 1 expression increased in high-flow and low-flow vessels, in association with a reduced mitochondrial aconitase activity (marker of oxidative stress) in high-flow arteries only. Inhibition of macrophage infiltration (clodronate) decreased heme oxygenase 1 induction in low-flow but not in high-flow arteries. Similarly, inhibition of NADPH oxidase activity (apocynin) decreased heme oxygenase 1 induction in low-flow but not high-flow arteries. However, dihydroethidium staining was higher in high-flow and low-flow compared with normal flow arteries. In arteries cannulated in an arteriograph, heme oxygenase 1 mRNA increased in a flow-dependent manner and was abolished by N(G)-nitro-l-arginine methyl ester, catalase, or mitochondrial electron transport chain inhibition. Furthermore, heme oxygenase 1 induction using cobalt-protoporphyrin restored altered high-flow remodeling in endothelial NO synthase knockout mice. Thus, in high-flow remodeling, heme oxygenase 1 induction depends on shear stress-generated NO and mitochondria-derived hydrogen peroxide. In low-flow remodeling, heme oxygenase 1 induction requires macrophage infiltration and is mediated by NADPH oxidase-derived superoxide.
Collapse
Affiliation(s)
- Mohamed Lamine Freidja
- Department of Integrated Neurovascular Biology, UMR CNRS 6214, INSERM U771, Faculté de Médecine, 49045 Angers, France
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Interleukin(IL)-4 promotion of CXCL-8 gene transcription is mediated by ERK1/2 pathway in human pulmonary artery endothelial cells. Mol Immunol 2011; 48:1784-92. [PMID: 21645924 DOI: 10.1016/j.molimm.2011.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 04/27/2011] [Accepted: 05/03/2011] [Indexed: 01/07/2023]
Abstract
Interleukin-4 is central to allergic pulmonary inflammatory responses, but its contribution to airway neutrophilia remains controversial. The endothelium plays a critical role in regulating leukocyte recruitment and migration during inflammation. However, its response to IL-4 is reported to either increase or decrease the production of neutrophil chemotactic factors. We hypothesized that these conflicting findings may be due to the tissue and the size of the vessels from which endothelial cells have been derived. The expression of CXCL-8 by human primary culture umbilical veins endothelial cells (HUVECs), human pulmonary artery endothelial cells (HPAECs), and human pulmonary microvascular endothelial cells (HPMECs) when stimulated with recombinant human IL-4 (rhIL-4) was studied. The chemoattractant property of the cells' supernatants for neutrophils was evaluated using Boyden chambers. The role of the nuclear factor-κB (NF-κB), and mitogen-activated protein kinases (MAPK) in IL-4-induced HPAECs was studied using Western blotting and electrophoretic mobility shift assay (EMSA). We demonstrated that IL-4 increased the mRNA expression and the protein production of CXCL-8 in HPAECs, but not in HUVECs and HPMECs. The supernatants of HAPECs stimulated by IL-4 significantly promoted neutrophils migration in a dose-dependent manner, and was significantly attenuated by an inhibitor of CXCL-8. We also found that extracellular-regulated protein kinase1/2 (ERK1/2) is activated by IL-4 in HPAECs, but not JUN-N-terminal protein kinase (JNK) or p38 MAPK pathway. Furthermore, NF-κB-DNA binding activity, phosphorylation of IκBα and p65 levels were not affected by rhIL-4 in HAPECs. These findings indicate marked functional differences in the response of micro and macro-ECs to IL-4. ERK1/2, rather than NF-κB, JNK and p38 MAPK signaling, plays a role in IL-4 induced chemokine activation. Our results suggest that inhibition of ERK1/2 may be a possible target for airway neutrophilia in allergic lung diseases.
Collapse
|
33
|
Malaguarnera G, Giordano M, Paladina I, Rando A, Uccello M, Basile F, Biondi A, Carnazzo S, Alessandria I, Mazzarino C. Markers of bile duct tumors. World J Gastrointest Oncol 2011; 3:49-59. [PMID: 21528090 PMCID: PMC3083496 DOI: 10.4251/wjgo.v3.i4.49] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 02/23/2011] [Accepted: 03/02/2011] [Indexed: 02/05/2023] Open
Abstract
Biliary tract carcinomas are relatively rare, representing less than 1% of cancers. However, their incidence has increased in Japan and in industrialized countries like the USA. Biliary tract tumors have a poor prognosis and a high mortality rate because they are usually detected late in the course of the disease; therapeutic treatment options are often limited and of minimal utility. Recent studies have shown the importance of serum and molecular markers in the diagnosis and follow up of biliary tract tumors. This review aims to introduce the main features of the most important serum and molecular markers of biliary tree tumors. Some considerable tumor markers are cancer antigen 125, carbohydrate antigen 19-9, carcinoembryonic antigen, chromogranin A, mucin 1, mucin 5, alpha-fetoprotein, claudins and cytokeratins.
Collapse
Affiliation(s)
- Giulia Malaguarnera
- Giulia Malaguarnera, Clorinda Mazzarino, Department of Biomedical Science, University of Catania, via Androne 83, 95124 Catania, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
It is becoming increasingly recognized that the host microenvironment is essential for regulating tumor cell behavior. The cellular stromal compartment can modulate angiogenesis either directly through enhanced secretion of pro-angiogenic factors or reduced secretion of antiangiogenic factors, or indirectly by modulating the surrounding extracellular matrix. Control of angiogenesis represents a critical step in cancer progression and is a potential therapeutic target. This article focuses on the role of the tumor microenvironment in the control of angiogenesis and how dissection of the molecular interactions may enhance prognostic and predictive power and facilitate therapeutic targeting.
Collapse
Affiliation(s)
- Sarah J L Payne
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | | |
Collapse
|
35
|
Chan YC, Khanna S, Roy S, Sen CK. miR-200b targets Ets-1 and is down-regulated by hypoxia to induce angiogenic response of endothelial cells. J Biol Chem 2010; 286:2047-56. [PMID: 21081489 DOI: 10.1074/jbc.m110.158790] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The miR-200 family plays a crucial role in epithelial to mesenchymal transition via controlling cell migration and polarity. We hypothesized that miR-200b, one miR-200 family member, could regulate angiogenic responses via modulating endothelial cell migration. Delivery of the miR-200b mimic in human microvascular endothelial cells (HMECs) suppressed the angiogenic response, whereas miR-200b-depleted HMECs exhibited elevated angiogenesis in vitro, as evidenced by Matrigel® tube formation and cell migration. Using in silico studies, miR target reporter assay, and Western blot analysis revealed that v-ets erythroblastosis virus E26 oncogene homolog 1 (Ets-1), a crucial angiogenesis-related transcription factor, serves as a novel direct target of miR-200b. Knocking down endogenous Ets-1 simulated an anti-angiogenic response of the miR-200b mimic-transfected cells. Certain Ets-1-associated genes, namely matrix metalloproteinase 1 and vascular endothelial growth factor receptor 2, were negatively regulated by miR-200b. Overexpression of Ets-1 rescued miR-200b-dependent impairment in angiogenic response and suppression of Ets-1-associated gene expression. Both hypoxia as well as HIF-1α stabilization inhibited miR-200b expression and elevated Ets-1 expression. Experiments to identify how miR-200b modulates angiogenesis under a low oxygen environment illustrated that hypoxia-induced miR-200b down-regulation de-repressed Ets-1 expression to promote angiogenesis. This study provides the first evidence that hypoxia-sensitive miR-200b is involved in induction of angiogenesis via directly targeting Ets-1 in HMECs.
Collapse
Affiliation(s)
- Yuk Cheung Chan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
36
|
Cui L, Qiao L, Wang J, Wang X, Yang Z, Ma B, Shi Y, Zhou X, Wu K, Han Y, Fan D. New Monoclonal Antibody B7 Selectively Recognizes Rat Myocardium Microvascular Endothelial Cells. Hybridoma (Larchmt) 2010; 29:413-8. [PMID: 21050042 DOI: 10.1089/hyb.2010.0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Lina Cui
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Lijuan Qiao
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jingbo Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xuechang Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhao Yang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Bin Ma
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xinmin Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ying Han
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
37
|
Siemianowicz K, Gminski J, Goss M, Francuz T, Likus W, Jurczak T, Garczorz W. Influence of elastin-derived peptides on metalloprotease production in endothelial cells. Exp Ther Med 2010; 1:1057-1060. [PMID: 22993640 DOI: 10.3892/etm.2010.157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 09/27/2010] [Indexed: 11/05/2022] Open
Abstract
Matrix metalloproteases (MMPs) are a family of zinc-dependent endopeptidases that degrade extracellular matrix proteins. MMP-1 and MMP-2 are produced by endothelial cells and are involved in specific vascular pathologies, including atherosclerosis and aortal aneurysm. One of the most important differences between these two metalloproteases is the possibility of hydrolysis of elastin and collagen type IV by MMP-2, but not by MMP-1. Elastin-derived peptides are generated as a result of the degradation of elastin fibers. The aim of our study was to compare the production of MMP-1 and MMP-2 in cultured human arterial endothelial cells derived from vascular pathologies localized at three different sites, the coronary artery, iliac artery and aorta, measured as their concentration in cell culture medium. The second aim was to evaluate the influence of κ-elastin (at concentrations 0.1, 0.4, 1.0, 2.5 or 5.0 μg/ml) on the production of the evaluated metalloproteases in three endothelial cell lines. The production of MMP-1 was statistically significantly greater in endothelial cells derived from the aorta compared to that in the endothelium obtained from the coronary and iliac arteries. There were no statistically significant differences in the production of MMP-2 among the endothelial cell lines tested. The addition of κ-elastin at all evaluated concentrations did not statistically significantly influence the concentration of MMP-1 in the cultured coronary artery endothelium. Furthermore, no statistically significant differences were observed in the cultured iliac artery endothelium. In the cultured endothelium derived from the aorta, κ-elastin at concentrations of 0.1 and 0.4 μg/ml significantly increased the amount of MMP-1.
Collapse
|
38
|
Ingthorsson S, Sigurdsson V, Fridriksdottir A, Jonasson JG, Kjartansson J, Magnusson MK, Gudjonsson T. Endothelial cells stimulate growth of normal and cancerous breast epithelial cells in 3D culture. BMC Res Notes 2010; 3:184. [PMID: 20609224 PMCID: PMC2909928 DOI: 10.1186/1756-0500-3-184] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 07/07/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Epithelial-stromal interaction provides regulatory signals that maintain correct histoarchitecture and homeostasis in the normal breast and facilitates tumor progression in breast cancer. However, research on the regulatory role of the endothelial component in the normal and malignant breast gland has largely been neglected. The aim of the study was to investigate the effects of endothelial cells on growth and differentiation of human breast epithelial cells in a three-dimensional (3D) co-culture assay. METHODS Breast luminal and myoepithelial cells and endothelial cells were isolated from reduction mammoplasties. Primary cells and established normal and malignant breast cell lines were embedded in reconstituted basement membrane in direct co-culture with endothelial cells and by separation of Transwell filters. Morphogenic and phenotypic profiles of co-cultures was evaluated by phase contrast microscopy, immunostaining and confocal microscopy. RESULTS In co-culture, endothelial cells stimulate proliferation of both luminal- and myoepithelial cells. Furthermore, endothelial cells induce a subpopulation of luminal epithelial cells to form large acini/ducts with a large and clear lumen. Endothelial cells also stimulate growth and cloning efficiency of normal and malignant breast epithelial cell lines. Transwell and gradient co-culture studies show that endothelial derived effects are mediated - at least partially - by soluble factors. CONCLUSION Breast endothelial cells - beside their role in transporting nutrients and oxygen to tissues - are vital component of the epithelial microenvironment in the breast and provide proliferative signals to the normal and malignant breast epithelium. These growth promoting effects of endothelial cells should be taken into consideration in breast cancer biology.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem cell research unit, Department of anatomy, Faculty of medicine, University of Iceland and Department of laboratory hematology, Landspitali, university hospital, (Vatnsmýrarvegur 16), Reykjavík, (101), Iceland.
| | | | | | | | | | | | | |
Collapse
|
39
|
Evaluation of endothelial cell culture as a model system of vascular ageing. Exp Gerontol 2010; 45:779-87. [PMID: 20600781 DOI: 10.1016/j.exger.2010.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 05/11/2010] [Accepted: 06/21/2010] [Indexed: 12/23/2022]
Abstract
The purpose of this study was to evaluate the relevance of long-term endothelial cell culture as a model system of vascular ageing. Micro- and macrovascular endothelial cells were serially passaged until replicative senescence and their ability to form tube-like structures when cultured on Matrigel was assessed throughout their lifespan. For both cell types low passage cultures adopted a homogeneous cobblestone morphology, while senescent cultures were extremely heterogeneous. Furthermore, both cell types showed a reduction in tube formation ability with in vitro ageing, which is in accordance with the reduction in angiogenic potential observed with ageing in vivo. Examination of senescence associated β-galactosidase activity revealed an increased activity in cells forming tubes as compared to cells cultured on plastic, which could be attributed to an increased lysosomal content of cells undergoing tube formation. As this increased senescence associated β-galactosidase activity was unrelated to the replicative age of the cells, senescence associated β-galactosidase activity may not be a relevant senescence marker for differentiating endothelial cells. The age-related reduction in tube formation ability suggested that long-term culture of endothelial cells may be a valid model system of vascular ageing, which makes it an ideal platform for high throughput screening of compounds influencing angiogenesis.
Collapse
|
40
|
Staton CA, Reed MWR, Brown NJ. A critical analysis of current in vitro and in vivo angiogenesis assays. Int J Exp Pathol 2009; 90:195-221. [PMID: 19563606 DOI: 10.1111/j.1365-2613.2008.00633.x] [Citation(s) in RCA: 344] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The study of angiogenesis has grown exponentially over the past 40 years with the recognition that angiogenesis is essential for numerous pathologies and, more recently, with the advent of successful drugs to inhibit angiogenesis in tumours. The main problem with angiogenesis research remains the choice of appropriate assays to evaluate the efficacy of potential new drugs and to identify potential targets within the angiogenic process. This selection is made more complex by the recognition that heterogeneity occurs, not only within the endothelial cells themselves, but also within the specific microenvironment to be studied. Thus, it is essential to choose the assay conditions and cell types that most closely resemble the angiogenic disease being studied. This is especially important when aiming to translate data from in vitro to in vivo and from preclinical to the clinic. Here we critically review and highlight recent advances in the principle assays in common use including those for endothelial cell proliferation, migration, differentiation and co-culture with fibroblasts and mural cells in vitro, vessel outgrowth from organ cultures and in vivo assays such as chick chorioallantoic membrane (CAM), zebrafish, sponge implantation, corneal, dorsal air sac, chamber and tumour angiogenesis models. Finally, we briefly discuss the direction likely to be taken in future studies, which include the use of increasingly sophisticated imaging analysis systems for data acquisition.
Collapse
Affiliation(s)
- Carolyn A Staton
- Microcirculation Research Group, Academic Unit of Surgical Oncology, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, UK.
| | | | | |
Collapse
|
41
|
McHarg S, Shore AC, Whatmore JL. Heterogeneity of Phospholipase D Activation by Angiotensin II, Lysophosphatidylcholine, and Insulin in Human Endothelial Cells. ACTA ACUST UNITED AC 2009; 15:213-8. [DOI: 10.1080/10623320802228765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Bruegmann E, Gruemmer R, Neulen J, Motejlek K. Regulation of soluble vascular endothelial growth factor receptor 1 secretion from human endothelial cells by tissue inhibitor of metalloproteinase 1. Mol Hum Reprod 2009; 15:749-56. [PMID: 19584194 DOI: 10.1093/molehr/gap053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) and its soluble receptor (sVEGFR-1) are key regulators in human ovarian angiogenesis. Produced by granulosa and ovarian theca interna cells, VEGF promotes blood vessel growth during follicular development and corpus luteum formation, whereas sVEGFR-1, which is secreted by endothelial cells, functions as an antagonist to VEGF activity by binding it. In order to gain further insights into the regulatory mechanisms of ovarian angiogenesis, the aim of the present study was to analyze the influence of tissue inhibitor of metalloproteinase 1 (TIMP-1), which is actively involved in the degradation and remodeling of the extracellular matrix, on sVEGFR-1 secretion of cultured human umbilical vein endothelial cells. sVEGFR-1 production was determined in the culture supernatant by Sandwich-ELISA. We showed that TIMP-1 produced by human granulosa cells and recombinant human TIMP-1 both significantly increased the production of sVEGFR-1 in endothelial cells. Also, the down-regulation of TIMP-1 expression by RNA interference resulted in a significant reduction of endothelial sVEGFR-1 secretion into the culture medium. Furthermore, TIMP-1 weakly inhibited proliferation of VEGF-stimulated endothelial cells. In conclusion, our results provide evidence that TIMP-1 increases the production of sVEGFR-1 in endothelial cells and thus may reduce VEGF bioavailability, leading to reduced blood vessel growth in the ovary.
Collapse
Affiliation(s)
- E Bruegmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | | |
Collapse
|
43
|
Abstract
Preceded by three decades of intense basic research on tumour angiogenesis, we are assisting to the translation of anti-antiangiogenic therapies as medical oncologists are increasingly using pioneering anti-angiogenic drugs in combination with standard treatments. While basic knowledge in the field of angiogenesis is reaching maturity and our level of understanding of the complex process of vessel development and growth in health and disease has been enriched at the molecular and cellular levels, the translation of this knowledge to the clinic is still in its infancy. Identifying the most suitable drugs, and the optimal dosage and schedule, as well as monitoring patients' responses to anti-angiogenic therapy, remain challenging issues that currently limit the benefit of this new therapeutic approach in cancer. This review will focus on a comprehensive description of the experimental assays in which angiogenesis research has been founded and how the different assays complement and provide relevant information for the task of characterising the angiogenic properties of diverse tumours, giving us a variety of tools to follow up tumour angiogenesis in research models. Following up tumour angiogenesis in patients and their response to antiangiogenic therapy is a more challenging task that will benefit in the near future from the use of non-invasive imaging methods as well as molecular and cellular biomarkers of angiogenesis suitable for clinical oncology. As both the design of the anti-angiogenic therapies and monitoring of the response are improved in the coming years to properly tailor them to the angiogenic profile of every patient, we hope to achieve increasing response and benefit of including antiangiogenic drugs as standard in cancer therapy.
Collapse
|
44
|
Yokoyama Y, Grünebach F, Schmidt SM, Heine A, Häntschel M, Stevanovic S, Rammensee HG, Brossart P. Matrilysin (MMP-7) is a novel broadly expressed tumor antigen recognized by antigen-specific T cells. Clin Cancer Res 2008; 14:5503-11. [PMID: 18765542 DOI: 10.1158/1078-0432.ccr-07-4041] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE A prerequisite for the development of vaccination strategies is the identification and characterization of relevant tumor-associated antigen. Using microarray and reverse transcription-PCR analysis, we found matrix metalloproteinase (MMP)-7 to be extensively up-regulated in renal cell carcinomas and expressed in a broad variety of malignant cells. MMP-7 can promote cancer invasion and angiogenesis by proteolytic cleavage of extracellular matrix and basement membrane proteins, thus making it a promising target in the context of immunotherapies. EXPERIMENTAL DESIGN To analyze the possible use of MMP-7 as a tumor-associated antigen, specific CTLs were induced using monocyte-derived dendritic cells electroporated with MMP-7-mRNA. In addition, to better characterize the fine specificity of these CTLs, MMP-7 MHC class I ligands were isolated and characterized in renal cell carcinoma tissue, which overexpressed MMP-7, by mass spectrometry-based peptide sequencing. Using this approach, we identified a novel HLA-A3-binding antigenic MMP-7 peptide. CTLs generated from healthy donors by in vitro priming with dendritic cells, pulsed with the novel peptide, were used as effectors in (51)Cr-release assays. RESULTS The induced CTLs elicited an antigen-specific and HLA-restricted cytolytic activity against tumor cells endogenously expressing the MMP-7 protein. Furthermore, we were able to induce MMP-7-specific CTLs using peripheral blood mononuclear cells from a patient with acute lymphoblastic leukemia capable of recognizing the autologous leukemic blasts while sparing nonmalignant cells. CONCLUSIONS Our study describes the identification of a novel broadly expressed T-cell epitope derived from the MMP-7 protein that represents an interesting candidate to be applied in immunotherapies of human malignancies targeting both tumor cells and neovascularization.
Collapse
Affiliation(s)
- Yuko Yokoyama
- Department Of Oncology, Hematology, Immunology, Rheumatology And Pulmology, University Of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hughes D, Fu AA, Puggioni A, Glockner JF, Anwer B, McGuire AM, Mukhopadhyay D, Misra S. Adventitial transplantation of blood outgrowth endothelial cells in porcine haemodialysis grafts alleviates hypoxia and decreases neointimal proliferation through a matrix metalloproteinase-9-mediated pathway--a pilot study. Nephrol Dial Transplant 2008; 24:85-96. [PMID: 18786975 PMCID: PMC2639314 DOI: 10.1093/ndt/gfn433] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose. We hypothesized that adventitial transplantation of blood outgrowth endothelial cells (BOEC) to the vein-to-graft anastomosis of polytetrafluoroethylene grafts will reduce neointimal hyperplasia by reducing hypoxia inducible factor-1α (HIF-1α), by increasing angiogenesis in a porcine model of chronic renal insufficiency with haemodialysis polytetrafluoroethylene grafts. Because matrix metalloproteinases (MMPs) have been shown to be involved with angiogenesis, the expression of MMPs and their inhibitors was determined. Methods. Chronic renal insufficiency was created by subtotal renal infarction and 28 days later, arteriovenous PTFE grafts were placed bilaterally from the carotid artery to the jugular vein. Autologous blood outgrowth endothelial cells labeled with Lac Z were transplanted to the adventitia of the vein-to-graft anastomosis using polyglycolic acid scaffolding and scaffolding only to other side (control). Animals were killed 14 days later and vessels were explanted from the vein-to-graft anastomosis of both sides and underwent immunohistochemical analysis, western blotting and zymography for HIF-1α, MMP-2, MMP-9, TIMP-1 and TIMP-2. BOEC were also made hypoxic and normoxic for 12, 24 and 48 h to determine protein expression for MMPs and TIMPs. Results. Under hypoxia, BOEC significantly increased the expression of pro MMP-2 by 12 h and TIMP-2 by 24 h when compared to normoxic cells (P < 0.05). Transplantation of BOEC resulted in a significant decrease in both HIF-1α and intima-to-media ratio with a significant increase in both pro and active MMP-9 when compared to control vessels (P < 0.05). MMP-9 activity was localized to the neointima of the transplanted vessels by immunohistochemistry. There was increased CD31 density with engraftment of BOEC cells into the neointima of both the transplanted vessels compared to controls (P = NS). Conclusion. Transplantation of BOEC resulted in a significant decrease in intimal hyperplasia and HIF-1α with a significant increase in both pro and active MMP-9 that was localized to the neointima of transplanted vessels. The increase in MMP-9 offers a possible mechanism for angiogenesis and the reduced intima-to-media ratio. Furthermore, we observed that BOEC had homed to the neointima of the contralateral vessels that had increased levels of HIF-1α, suggesting that hypoxia may be an important stimulus for BOEC migration.
Collapse
Affiliation(s)
- Deborah Hughes
- Division of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Expression of MMP-9 and TIMP-1 in lesions of systemic sclerosis and its implications. ACTA ACUST UNITED AC 2008; 28:480-2. [DOI: 10.1007/s11596-008-0424-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Indexed: 11/26/2022]
|
47
|
De Filippis D, D'Amico A, Cinelli MP, Esposito G, Di Marzo V, Iuvone T. Adelmidrol, a palmitoylethanolamide analogue, reduces chronic inflammation in a carrageenin-granuloma model in rats. J Cell Mol Med 2008; 13:1086-95. [PMID: 18429935 PMCID: PMC4496105 DOI: 10.1111/j.1582-4934.2008.00353.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Palmitoylethanolamide (PEA) and some of its analogues have shown great efficacy in the treatment of pain and inflammation. Adelmidrol – the International Nonproprietary Name (INN) of the di-amide derivative of azelaic acid – is one of these analogues. The anti-inflammatory and analgesic effects of PEA and adelmidrol are hypothesized to be mediated, at least in part, by mast cell down-modulation. Mast cell mediators released at early stage of the inflammatory process drive the inflammatory reaction to chronicity as it happens in X-carrageenin-induced granulomatous tissue formation. In the present study, the choice of testing adelmidrol depends upon the physicochemical properties of the compound, i.e. the amphipatic feature, that make it more easily soluble than PEA. In this study, we investigated the effect of adelmidrol on granuloma formation induced by λ-carrageenin-soaked sponge implant in rats. Our results show that the local administration of the compound under study significantly decreases weight and neo-angiogenesis in granulomatous tissue. The anti-inflammatory effect was due to the modulation of mast cells degranulation, as shown by histological analysis and by the inhibition of the release of several pro-inflammatory and pro-angiogenic enzymes (e.g. iNOS, chymase and metalloproteinase MMP-9), and mediators (e.g. nitric oxide and TNF-α). The results indicate that adelmidrol, given locally, may represent a potential therapeutic tool in controlling chronic inflammation.
Collapse
|
48
|
|
49
|
Bakos RM, Bakos L, Albano Edelweiss MI, Cartell A, Mariante JC, Soles Masiero NCM. Immunohistochemical expression of matrix metalloproteinase-2 and -9 in melanocytic nevi is altered by ultraviolet B. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2007; 23:250-4. [DOI: 10.1111/j.1600-0781.2007.00322.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Goodwin AM. In vitro assays of angiogenesis for assessment of angiogenic and anti-angiogenic agents. Microvasc Res 2007; 74:172-83. [PMID: 17631914 PMCID: PMC2692317 DOI: 10.1016/j.mvr.2007.05.006] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Revised: 05/02/2007] [Accepted: 05/10/2007] [Indexed: 12/27/2022]
Abstract
Blood vessels, either in insufficient numbers or in excess, contribute to the pathogenesis of many diseases. Agents that stimulate angiogenesis can improve blood flow in patients with ischemic diseases, whereas anti-angiogenic agents are used to treat disorders ranging from macular degeneration to cancer. In this review I describe in vitro assays that can be used to assess the activity of agents that affect angiogenesis. Means of quantifying endothelial cell matrix degradation, migration, proliferation, apoptosis and morphogenesis are discussed, as are embryoid body, aortic ring and metatarsal assays of vessel outgrowth. Strengths and limitations of these techniques are also addressed.
Collapse
Affiliation(s)
- Anne M Goodwin
- Department of Biology, Massachusetts College of Liberal Arts, 375 Church St., North Adams, MA 01247, USA.
| |
Collapse
|