1
|
Inafuku N, Sowa Y, Kishida T, Sawai S, Ntege EH, Numajiri T, Yamamoto K, Shimizu Y, Mazda O. Investigation of the stemness and wound-healing potential of long-term cryopreserved stromal vascular fraction cells. Regen Ther 2025; 29:128-139. [PMID: 40162021 PMCID: PMC11952815 DOI: 10.1016/j.reth.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Stromal vascular fraction (SVF), a heterogeneous cell population primarily derived from adipose tissue, is widely utilized in regenerative therapies for its wound-healing properties and accessibility. While its immediate availability is advantageous, repeated harvesting can be burdensome, especially for elderly patients, and the regenerative capacity of SVF declines with donor age. Long-term cryopreservation offers a potential solution by allowing the banking of SVF from younger donors for future use; however, the impact of this process on SVF functionality remains elusive. This study investigates the stemness and wound-healing potential of SVF following prolonged cryopreservation. Methods SVF cells were isolated from adipose tissue harvested from twelve patients and cryopreserved for either two months (short-term cryopreserved SVF, S-SVF) or 12-13 years (long-term cryopreserved SVF, L-SVF), with six patients in each group. In vitro assays assessed cell viability and stemness, while in vivo assays evaluated wound-healing ability by administering thawed SVF cells from each group to dorsal wounds in immunodeficient mice, compared with a control group. Non-parametric statistical tests analyzed the differences between groups. Results L-SVF exhibited significantly lower stemness compared to S-SVF. Importantly, the L-SVF group showed significantly improved wound healing compared with the control group, although the wound-healing effect of L-SVF was inferior to that of the S-SVF. Conclusion This study demonstrated that, despite reduced stemness, L-SVF retains partial wound-healing potential after 12-13 years of cryopreservation. These findings highlight the need for optimized cryopreservation protocols to enhance SVF viability and regenerative capacity for clinical applications.
Collapse
Affiliation(s)
- Naoki Inafuku
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Seiji Sawai
- Department of Orthopedics, Jyujyo Takeda Rehabilitation Hospital, Kyoto, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Toshiaki Numajiri
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Kenta Yamamoto
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo, Kyoto, Japan
| |
Collapse
|
2
|
Huang YH, Watanabe M, Yamashita T, Sudo R. Construction of highly vascularized hepatic spheroids of primary hepatocytes via pro-angiogenic strategy in vitro. Biofabrication 2025; 17:035001. [PMID: 40179891 DOI: 10.1088/1758-5090/adc8d4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/03/2025] [Indexed: 04/05/2025]
Abstract
Primary hepatocytes are widely recognized for their ability to accurately represent thein vivohepatocyte phenotype. However, traditional avascular primary hepatocyte culture models are limited by inadequate mass transfer, which leads to a rapid decline in hepatocyte function and survival. To address these challenges, vascularization of hepatic spheroids is crucial for enhancing oxygen and nutrient supply, thereby enabling the construction of larger and more complex hepatic tissuesin vitro. In this study, we achieved vascularization of hepatic spheroids containing freshly isolated primary hepatocytes by incorporating fibroblasts as a source of paracrine factors to induce angiogenesis. Multicellular spheroids composed of primary hepatocytes and fibroblasts were formed in non-adhesive concave wells, and one of the spheroids was subsequently embedded in a fibrin-collagen hydrogel within a microfluidic device. Endothelial cells were then seeded onto adjacent microfluidic channels. They formed microvascular networks that extended toward and penetrated the hepatic spheroid. The vascularized hepatic spheroid closely mimicked hepatic sinusoids, with hepatocytes in close contact with microvessels. Moreover, the vascularized spheroid exhibited significantly enhanced hepatic function, specifically albumin secretion and urea synthesis. Our findings provide insights into the establishment of highly vascularized hepatic spheroidsin vitro, which is crucial for constructing scalable hepatic tissues in the context of biofabrication.
Collapse
Affiliation(s)
- Yen-Hsiang Huang
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Masafumi Watanabe
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| | - Tadahiro Yamashita
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| | - Ryo Sudo
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| |
Collapse
|
3
|
Landau S, Okhovatian S, Zhao Y, Liu C, Shakeri A, Wang Y, Ramsay K, Kieda J, Jiang R, Radisic M. Bioengineering vascularization. Development 2024; 151:dev204455. [PMID: 39611864 PMCID: PMC11698057 DOI: 10.1242/dev.204455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
This Review explores the rapidly evolving field of bioengineered vasculature, a key area of focus in tissue engineering and regenerative medicine. The broad relevance of this topic is attributed to its impacts on a wide range of biological processes, enabling studies in tissue development, fundamental biology and drug discovery, and the applications in tissue engineering and regenerative medicine. We outline the design criteria for bioengineered vasculature and the methodologies for constructing these systems by self-assembly and in microfluidics, organs-on-a-chip and macroscale tubular systems that often rely on biofabrication approaches such as 3D printing. We discuss existing challenges in developing functional vasculature that closely mirrors its native equivalent, including achieving hierarchical branching with organ and vessel-specific endothelial and supporting cells, providing perusable vasculature within organoids and scaling the systems for implantation and direct vascular anastomosis.
Collapse
Affiliation(s)
- Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto M5G 1X6, ON, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Kaitlyn Ramsay
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto M5S 3E5, ON, Canada
- Terence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, ON, Canada
| |
Collapse
|
4
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
5
|
Murphy AR, Allenby MC. In vitro microvascular engineering approaches and strategies for interstitial tissue integration. Acta Biomater 2023; 171:114-130. [PMID: 37717711 DOI: 10.1016/j.actbio.2023.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
The increasing gap between clinical demand for tissue or organ transplants and the availability of donated tissue highlights the emerging opportunities for lab-grown or synthetically engineered tissue. While the field of tissue engineering has existed for nearly half a century, its clinical translation remains unrealised, in part, due to a limited ability to engineer sufficient vascular supply into fabricated tissue, which is necessary to enable nutrient and waste exchange, prevent cellular necrosis, and support tissue proliferation. Techniques to develop anatomically relevant, functional vascular networks in vitro have made significant progress in the last decade, however, the challenge now remains as to how best incorporate these throughout dense parenchymal tissue-like structures to address diffusion-limited development and allow for the fabrication of large-scale vascularised tissue. This review explores advances made in the laboratory engineering of vasculature structures and summarises recent attempts to integrate vascular networks together with sophisticated in vitro avascular tissue and organ-like structures. STATEMENT OF SIGNIFICANCE: The ability to grow full scale, functional tissue and organs in vitro is primarily limited by an inability to adequately diffuse oxygen and nutrients throughout developing cellularised structures, which generally results from the absence of perfusable vessel networks. Techniques to engineering both perfusable vascular networks and avascular miniaturised organ-like structures have recently increased in complexity, sophistication, and physiological relevance. However, integrating these two essential elements into a single functioning vascularised tissue structure represents a significant spatial and temporal engineering challenge which is yet to be surmounted. Here, we explore a range of vessel morphogenic phenomena essential for tissue-vascular co-development, as well as evaluate a range of recent noteworthy approaches for generating vascularised tissue products in vitro.
Collapse
Affiliation(s)
- A R Murphy
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia
| | - M C Allenby
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia; Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
6
|
Curtis MB, Kelly N, Hughes CCW, George SC. Organotypic stromal cells impact endothelial cell transcriptome in 3D microvessel networks. Sci Rep 2022; 12:20434. [PMID: 36443378 PMCID: PMC9705391 DOI: 10.1038/s41598-022-24013-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Endothelial cells line all major blood vessels and serve as integral regulators of many functions including vessel diameter, cellular trafficking, and transport of soluble mediators. Despite similar functions, the phenotype of endothelial cells is highly organ-specific, yet our understanding of the mechanisms leading to organ-level differentiation is incomplete. We generated 3D microvessel networks by combining a common naïve endothelial cell with six different stromal cells derived from the lung, skin, heart, bone marrow, pancreas, and pancreatic cancer. Single cell RNA-Seq analysis of the microvessel networks reveals five distinct endothelial cell populations, for which the relative proportion depends on the stromal cell population. Morphologic features of the organotypic vessel networks inversely correlate with a cluster of endothelial cells associated with protein synthesis. The organotypic stromal cells were each characterized by a unique subpopulation of cells dedicated to extracellular matrix organization and assembly. Finally, compared to cells in 2D monolayer, the endothelial cell transcriptome from the 3D in vitro heart, skin, lung, and pancreas microvessel networks are more similar to the in vivo endothelial cells from the respective organs. We conclude that stromal cells contribute to endothelial cell and microvessel network organ tropism, and create an endothelial cell phenotype that more closely resembles that present in vivo.
Collapse
Affiliation(s)
- Matthew B Curtis
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA
| | - Natalie Kelly
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA.
| |
Collapse
|
7
|
Miwa H, Dimatteo R, de Rutte J, Ghosh R, Di Carlo D. Single-cell sorting based on secreted products for functionally defined cell therapies. MICROSYSTEMS & NANOENGINEERING 2022; 8:84. [PMID: 35874174 PMCID: PMC9303846 DOI: 10.1038/s41378-022-00422-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 05/13/2023]
Abstract
Cell therapies have emerged as a promising new class of "living" therapeutics over the last decade and have been particularly successful for treating hematological malignancies. Increasingly, cellular therapeutics are being developed with the aim of treating almost any disease, from solid tumors and autoimmune disorders to fibrosis, neurodegenerative disorders and even aging itself. However, their therapeutic potential has remained limited due to the fundamental differences in how molecular and cellular therapies function. While the structure of a molecular therapeutic is directly linked to biological function, cells with the same genetic blueprint can have vastly different functional properties (e.g., secretion, proliferation, cell killing, migration). Although there exists a vast array of analytical and preparative separation approaches for molecules, the functional differences among cells are exacerbated by a lack of functional potency-based sorting approaches. In this context, we describe the need for next-generation single-cell profiling microtechnologies that allow the direct evaluation and sorting of single cells based on functional properties, with a focus on secreted molecules, which are critical for the in vivo efficacy of current cell therapies. We first define three critical processes for single-cell secretion-based profiling technology: (1) partitioning individual cells into uniform compartments; (2) accumulating secretions and labeling via reporter molecules; and (3) measuring the signal associated with the reporter and, if sorting, triggering a sorting event based on these reporter signals. We summarize recent academic and commercial technologies for functional single-cell analysis in addition to sorting and industrial applications of these technologies. These approaches fall into three categories: microchamber, microfluidic droplet, and lab-on-a-particle technologies. Finally, we outline a number of unmet needs in terms of the discovery, design and manufacturing of cellular therapeutics and how the next generation of single-cell functional screening technologies could allow the realization of robust cellular therapeutics for all patients.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Joseph de Rutte
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Partillion Bioscience, Los Angeles, CA 90095 USA
| | - Rajesh Ghosh
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
8
|
Seymour AJ, Westerfield AD, Cornelius VC, Skylar-Scott MA, Heilshorn SC. Bioprinted microvasculature: progressing from structure to function. Biofabrication 2022; 14:10.1088/1758-5090/ac4fb5. [PMID: 35086069 PMCID: PMC8988885 DOI: 10.1088/1758-5090/ac4fb5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/27/2022] [Indexed: 11/12/2022]
Abstract
Three-dimensional (3D) bioprinting seeks to unlock the rapid generation of complex tissue constructs, but long-standing challenges with efficientin vitromicrovascularization must be solved before this can become a reality. Microvasculature is particularly challenging to biofabricate due to the presence of a hollow lumen, a hierarchically branched network topology, and a complex signaling milieu. All of these characteristics are required for proper microvascular-and, thus, tissue-function. While several techniques have been developed to address distinct portions of this microvascularization challenge, no single approach is capable of simultaneously recreating all three microvascular characteristics. In this review, we present a three-part framework that proposes integration of existing techniques to generate mature microvascular constructs. First, extrusion-based 3D bioprinting creates a mesoscale foundation of hollow, endothelialized channels. Second, biochemical and biophysical cues induce endothelial sprouting to create a capillary-mimetic network. Third, the construct is conditioned to enhance network maturity. Across all three of these stages, we highlight the potential for extrusion-based bioprinting to become a central technique for engineering hierarchical microvasculature. We envision that the successful biofabrication of functionally engineered microvasculature will address a critical need in tissue engineering, and propel further advances in regenerative medicine andex vivohuman tissue modeling.
Collapse
Affiliation(s)
- Alexis J. Seymour
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Ashley D. Westerfield
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Vincent C. Cornelius
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Mark A. Skylar-Scott
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science & Engineering, Stanford University, 476 Lomita Mall, McCullough Room 246, Stanford, CA 94305, USA
| |
Collapse
|
9
|
Walji N, Kheiri S, Young EWK. Angiogenic Sprouting Dynamics Mediated by Endothelial-Fibroblast Interactions in Microfluidic Systems. Adv Biol (Weinh) 2021; 5:e2101080. [PMID: 34655165 DOI: 10.1002/adbi.202101080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/18/2021] [Indexed: 11/09/2022]
Abstract
Angiogenesis, the development of new blood vessels from existing vasculature, is a key process in normal development and pathophysiology. In vitro models are necessary for investigating mechanisms of angiogenesis and developing antiangiogenic therapies. Microfluidic cell culture models of angiogenesis are favored for their ability to recapitulate 3D tissue structures and control spatiotemporal aspects of the microenvironments. To capture the angiogenesis process, microfluidic models often include endothelial cells and a fibroblast component. However, the influence of fibroblast organization on resulting angiogenic behavior remains unclear. Here a comparative study of angiogenic sprouting on a microfluidic chip induced by fibroblasts in 2D monolayer, 3D dispersed, and 3D spheroid culture formats, is conducted. Vessel morphology and sprout distribution for each configuration are measured, and these observations are correlated with measurements of secreted factors and numerical simulations of diffusion gradients. The results demonstrate that angiogenic sprouting varies in response to fibroblast organization with correlating variations in secretory profile and secreted factor gradients across the microfluidic device. This study is anticipated to shed light on how sprouting dynamics are mediated by fibroblast configuration such that the microfluidic cell culture design process includes the selection of a fibroblast component where the effects are known and leveraged.
Collapse
Affiliation(s)
- Noosheen Walji
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| | - Sina Kheiri
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada
| | - Edmond W K Young
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| |
Collapse
|
10
|
Browne S, Gill EL, Schultheiss P, Goswami I, Healy KE. Stem cell-based vascularization of microphysiological systems. Stem Cell Reports 2021; 16:2058-2075. [PMID: 33836144 PMCID: PMC8452487 DOI: 10.1016/j.stemcr.2021.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Microphysiological systems (MPSs) (i.e., tissue or organ chips) exploit microfluidics and 3D cell culture to mimic tissue and organ-level physiology. The advent of human induced pluripotent stem cell (hiPSC) technology has accelerated the use of MPSs to study human disease in a range of organ systems. However, in the reduction of system complexity, the intricacies of vasculature are an often-overlooked aspect of MPS design. The growing library of pluripotent stem cell-derived endothelial cell and perivascular cell protocols have great potential to improve the physiological relevance of vasculature within MPS, specifically for in vitro disease modeling. Three strategic categories of vascular MPS are outlined: self-assembled, interface focused, and 3D biofabricated. This review discusses key features and development of the native vasculature, linking that to how hiPSC-derived vascular cells have been generated, the state of the art in vascular MPSs, and opportunities arising from interdisciplinary thinking.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Elisabeth L Gill
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Paula Schultheiss
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
11
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
12
|
Song HHG, Lammers A, Sundaram S, Rubio L, Chen AX, Li L, Eyckmans J, Bhatia SN, Chen CS. Transient Support from Fibroblasts is Sufficient to Drive Functional Vascularization in Engineered Tissues. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2003777. [PMID: 33613149 PMCID: PMC7891457 DOI: 10.1002/adfm.202003777] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 05/05/2023]
Abstract
Formation of capillary blood vasculature is a critical requirement for native as well as engineered organs and can be induced in vitro by co-culturing endothelial cells with fibroblasts. However, whether these fibroblasts are required only in the initial morphogenesis of endothelial cells or needed throughout is unknown, and the ability to remove these stromal cells after assembly could be useful for clinical translation. In this study, we introduce a technique termed CAMEO (Controlled Apoptosis in Multicellular Tissues for Engineered Organogenesis), whereby fibroblasts are selectively ablated on demand, and utilize it to probe the dispensability of fibroblasts in vascular morphogenesis. The presence of fibroblasts is shown to be necessary only during the first few days of endothelial cell morphogenesis, after which they can be ablated without significantly affecting the structural and functional features of the developed vasculature. Furthermore, we demonstrate the use of CAMEO to vascularize a construct containing primary human hepatocytes that improved tissue function. In conclusion, this study suggests that transient, initial support from fibroblasts is sufficient to drive vascular morphogenesis in engineered tissues, and this strategy of engineering-via-elimination may provide a new general approach for achieving desired functions and cell compositions in engineered organs.
Collapse
Affiliation(s)
- H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alex Lammers
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Subramanian Sundaram
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Logan Rubio
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Linqing Li
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jeroen Eyckmans
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
13
|
Sugihara K, Yamaguchi Y, Usui S, Nashimoto Y, Hanada S, Kiyokawa E, Uemura A, Yokokawa R, Nishiyama K, Miura T. A new perfusion culture method with a self-organized capillary network. PLoS One 2020; 15:e0240552. [PMID: 33112918 PMCID: PMC7592787 DOI: 10.1371/journal.pone.0240552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/28/2020] [Indexed: 01/06/2023] Open
Abstract
A lack of perfusion has been one of the most significant obstacles for three-dimensional culture systems of organoids and embryonic tissues. Here, we developed a simple and reliable method to implement a perfusable capillary network in vitro. The method employed the self-organization of endothelial cells to generate a capillary network and a static pressure difference for culture medium circulation, which can be easily introduced to standard biological laboratories and enables long-term cultivation of vascular structures. Using this culture system, we perfused the lumen of the self-organized capillary network and observed a flow-induced vascular remodeling process, cell shape changes, and collective cell migration. We also observed an increase in cell proliferation around the self-organized vasculature induced by flow, indicating functional perfusion of the culture medium. We also reconstructed extravasation of tumor and inflammatory cells, and circulation inside spheroids including endothelial cells and human lung fibroblasts. In conclusion, this system is a promising tool to elucidate the mechanisms of various biological processes related to vascular flow.
Collapse
Affiliation(s)
- Kei Sugihara
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshimi Yamaguchi
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shiori Usui
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuji Nashimoto
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Department of Micro Engineering, Kyoto University, Kyoto, Japan
| | - Sanshiro Hanada
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Etsuko Kiyokawa
- Department of Oncologic Pathology, Kanazawa Medical University, Ishikawa, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, Japan
| | - Koichi Nishiyama
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Takashi Miura
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
14
|
Zhao X, Guo J, Zhang F, Zhang J, Liu D, Hu W, Yin H, Jin L. Therapeutic application of adipose-derived stromal vascular fraction in diabetic foot. Stem Cell Res Ther 2020; 11:394. [PMID: 32928305 PMCID: PMC7488783 DOI: 10.1186/s13287-020-01825-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/15/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic foot is one of the severest complications of diabetes. In severe cases, this disease may be lead to amputation or even death due to secondary infection and ischemic necrosis. Since the ineffectiveness of traditional therapy, autologous stem cell transplantation has been used to treat diabetic foot. This simple, safe, and effective therapy is expected to be applied and promoted in the future.In this review, we described the detailed pathogenesis of diabetic foot and the common clinical treatments currently used. We also revealed vascular remodeling as the potential mechanism of therapeutic functions of adipose-derived stromal vascular fraction (SVF) in treating diabetic foot.
Collapse
Affiliation(s)
- Xiansheng Zhao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China
| | - Jiamin Guo
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, California, 91010, USA
| | - Fangfang Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China
| | - Jue Zhang
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Delin Liu
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Wenjun Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China.
| | - Han Yin
- Department of Endocrinology, The Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.
| | - Liang Jin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu Province, China.
| |
Collapse
|
15
|
Rambøl MH, Han E, Niklason LE. Microvessel Network Formation and Interactions with Pancreatic Islets in Three-Dimensional Chip Cultures. Tissue Eng Part A 2020; 26:556-568. [PMID: 31724494 PMCID: PMC7249478 DOI: 10.1089/ten.tea.2019.0186] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
The pancreatic islet is a highly vascularized micro-organ, and rapid revascularization postislet transplantation is important for islet survival and function. However, the various mechanisms involved in islet revascularization are not fully understood, and we currently lack good in vitro platforms to explore this. Our aim for this study was to generate perfusable microvascular networks in a microfluidic chip device, in which islets could be easily integrated, to establish an in vitro platform for investigations on islet-microvasculature interactions. We compared the ability of mesenchymal stem cells (MSCs) and fibroblasts to support microvascular network formation by human umbilical vein endothelial cells (HUVECs) and human induced pluripotent stem cell-derived endothelial colony-forming cell in two-dimensional and three-dimensional models of angiogenesis, and tested the effect of different culture media on microvessel formation. HUVECs that were supported by MSCs formed patent and perfusable networks in a fibrin gel, whereas networks supported by fibroblasts rapidly regressed. Network morphology could be controlled by adjusting relative cell numbers and densities. Incorporation of isolated rat islets demonstrated that islets recruit local microvasculature in vitro, but that the microvessels did not invade islets, at least during the course of these studies. This in vitro microvascularization platform can provide a useful tool to study how various parameters affect islet integration with microvascular networks and could also be utilized for studies of vascularization of other organ systems. Impact statement To improve pancreatic islet graft survival and function posttransplantation, rapid and adequate revascularization is critical. Efforts to improve islet revascularization are demanding due to an insufficient understanding of the mechanisms involved in the process. We have applied a microfluidics platform to generate microvascular networks, and by incorporating pancreatic islets, we were able to study microvasculature-islet interactions in real time. This platform can provide a useful tool to study islet integration with microvascular networks, and could be utilized for studies of vascularization of other organ systems. Moreover, this work may be adapted toward developing a prevascularized islet construct for transplantation.
Collapse
Affiliation(s)
- Mia H. Rambøl
- Department of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Edward Han
- Department of Biomedical Engineering and Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| | - Laura E. Niklason
- Department of Biomedical Engineering and Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Kosyakova N, Kao DD, Figetakis M, López-Giráldez F, Spindler S, Graham M, James KJ, Won Shin J, Liu X, Tietjen GT, Pober JS, Chang WG. Differential functional roles of fibroblasts and pericytes in the formation of tissue-engineered microvascular networks in vitro. NPJ Regen Med 2020; 5:1. [PMID: 31934351 PMCID: PMC6944695 DOI: 10.1038/s41536-019-0086-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Formation of a perfusable microvascular network (μVN) is critical for tissue engineering of solid organs. Stromal cells can support endothelial cell (EC) self-assembly into a μVN, but distinct stromal cell populations may play different roles in this process. Here we describe the differential effects that two widely used stromal cell populations, fibroblasts (FBs) and pericytes (PCs), have on μVN formation. We examined the effects of adding defined stromal cell populations on the self-assembly of ECs derived from human endothelial colony forming cells (ECFCs) into perfusable μVNs in fibrin gels cast within a microfluidic chamber. ECs alone failed to fully assemble a perfusable μVN. Human lung FBs stimulated the formation of EC-lined μVNs within microfluidic devices. RNA-seq analysis suggested that FBs produce high levels of hepatocyte growth factor (HGF). Addition of recombinant HGF improved while the c-MET inhibitor, Capmatinib (INCB28060), reduced μVN formation within devices. Human placental PCs could not substitute for FBs, but in the presence of FBs, PCs closely associated with ECs, formed a common basement membrane, extended microfilaments intercellularly, and reduced microvessel diameters. Different stromal cell types provide different functions in microvessel assembly by ECs. FBs support μVN formation by providing paracrine growth factors whereas PCs directly interact with ECs to modify microvascular morphology.
Collapse
Affiliation(s)
- Natalia Kosyakova
- Department of Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Derek D. Kao
- Yale College of Undergraduate Studies, Yale University, New Haven, CT 06520 USA
| | - Maria Figetakis
- Department of Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520 USA
| | | | - Susann Spindler
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Morven Graham
- Yale Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Kevin J. James
- Department of Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Jee Won Shin
- Yale College of Undergraduate Studies, Yale University, New Haven, CT 06520 USA
| | - Xinran Liu
- Yale Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Gregory T. Tietjen
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Jordan S. Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519 USA
| | - William G. Chang
- Department of Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520 USA
| |
Collapse
|
17
|
Ribeiro TO, Silveira BM, Meira MC, Carreira ACO, Sogayar MC, Meyer R, Fortuna V. Investigating the potential of the secretome of mesenchymal stem cells derived from sickle cell disease patients. PLoS One 2019; 14:e0222093. [PMID: 31665139 PMCID: PMC6821040 DOI: 10.1371/journal.pone.0222093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Sickle cell disease (SCD) is a monogenic red cell disorder associated with multiple vascular complications, microvessel injury and wound-healing deficiency. Although stem cell transplantation with bone marrow-derived mesenchymal stem cells (BMSC) can promote wound healing and tissue repair in SCD patients, therapeutic efficacy is largely dependent on the paracrine activity of the implanted BM stromal cells. Since in vitro expansion and culture conditions are known to modulate the innate characteristics of BMSCs, the present study investigated the effects of normoxic and hypoxic cell-culture preconditioning on the BMSC secretome, in addition to the expression of paracrine molecules that induce angiogenesis and skin regeneration. BMSCs derived from SCD patients were submitted to culturing under normoxic (norCM) and hypoxic (hypoCM) conditions. We found that hypoxically conditioned cells presented increased expression and secretion of several well-characterized trophic growth factors (VEGF, IL8, MCP-1, ANG) directly linked to angiogenesis and tissue repair. The hypoCM secretome presented stronger angiogenic potential than norCM, both in vitro and in vivo, as evidenced by HUVEC proliferation, survival, migration, sprouting formation and in vivo angiogenesis. After local application in a murine wound-healing model, HypoCM showed significantly improved wound closure, as well as enhanced neovascularization in comparison to untreated controls. In sum, the secretome of hypoxia-preconditioned BMSC has increased expression of trophic factors involved in angiogenesis and skin regeneration. Considering that these preconditioned media are easily obtainable, this strategy represents an alternative to stem cell transplantation and could form the basis of novel therapies for vascular regeneration and wound healing in individuals with sickle cell disease.
Collapse
Affiliation(s)
- Tiago O. Ribeiro
- Health Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| | - Brysa M. Silveira
- Health Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| | - Mercia C. Meira
- Health Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| | - Ana C. O. Carreira
- Cell and Molecular Therapy Center NUCEL-NETCEM, School of Medicine, Internal Medicine Department, University of São Paulo, São Paulo, SP, Brazil
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center NUCEL-NETCEM, School of Medicine, Internal Medicine Department, University of São Paulo, São Paulo, SP, Brazil
- Chemistry Institute, Biochemistry Department, University of São Paulo, São Paulo, SP, Brazil
| | - Roberto Meyer
- Health Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| | - Vitor Fortuna
- Health Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| |
Collapse
|
18
|
Abe Y, Watanabe M, Chung S, Kamm RD, Tanishita K, Sudo R. Balance of interstitial flow magnitude and vascular endothelial growth factor concentration modulates three-dimensional microvascular network formation. APL Bioeng 2019; 3:036102. [PMID: 31431938 PMCID: PMC6697031 DOI: 10.1063/1.5094735] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/09/2019] [Indexed: 12/02/2022] Open
Abstract
Hemodynamic and biochemical factors play important roles in critical steps of angiogenesis. In particular, interstitial flow has attracted attention as an important hemodynamic factor controlling the angiogenic process. Here, we applied a wide range of interstitial flow magnitudes to an in vitro three-dimensional (3D) angiogenesis model in a microfluidic device. This study aimed to investigate the effect of interstitial flow magnitude in combination with the vascular endothelial growth factor (VEGF) concentration on 3D microvascular network formation. Human umbilical vein endothelial cells (HUVECs) were cultured in a series of interstitial flow generated by 2, 8, and 25 mmH2O. Our findings indicated that interstitial flow significantly enhanced vascular sprout formation, network extension, and the development of branching networks in a magnitude-dependent manner. Furthermore, we demonstrated that the proangiogenic effect of interstitial flow application could not be substituted by the increased VEGF concentration. In addition, we found that HUVECs near vascular sprouts significantly elongated in >8 mmH2O conditions, while activation of Src was detected even in 2 mmH2O conditions. Our results suggest that the balance between the interstitial flow magnitude and the VEGF concentration plays an important role in the regulation of 3D microvascular network formation in vitro.
Collapse
Affiliation(s)
- Yoshinori Abe
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Masafumi Watanabe
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul 02841, South Korea
| | - Roger D Kamm
- Departments of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Boston, Massachusetts 02139, USA
| | - Kazuo Tanishita
- Department of System Design Engineering, Keio University, Yokohama 223-8522, Japan
| | | |
Collapse
|
19
|
Khalid RS, Khan I, Zaidi MB, Naeem N, Haneef K, Qazi REM, Habib R, Malick TS, Ali A, Salim A. IL-7 overexpression enhances therapeutic potential of rat bone marrow mesenchymal stem cells for diabetic wounds. Wound Repair Regen 2019; 27:235-248. [PMID: 30761686 DOI: 10.1111/wrr.12706] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 02/11/2019] [Indexed: 12/28/2022]
Abstract
This study was aimed to enhance the healing potential of rat bone marrow mesenchymal stem cells against chronic diabetic wounds through interleukin-7 (IL-7) transfection. IL-7 plays an important role in wound healing and acts as a survival factor in some cell types. This study involves isolation, propagation, and characterization of mesenchymal stem cells (MSCs) and their modification with IL-7 gene via retroviral transfection. Transfected MSCs were assessed for their effect on angiogenic genes by qPCR. Wound healing potential of transfected MSCs was analyzed by scratch assay in vitro and by transplanting these cells in rat diabetic wound models in vivo. Wound area was measured for a period of 15 days and subsequent histological analysis was performed. qPCR results showed increased expression of IL-7 gene (p ≤ 0.05) and also principal angiogenic genes, vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), VEGF receptor 1 (FLT-1), and VEGF receptor 2 (FLK-1) (p ≤ 0.05). Neuropilin-1 (NRP-1) did not show any significant change. In vitro analysis of IL-7 MSCs showed intense cell-cell connections and tube formation as compared to the normal MSCs. Rate of wound closure was more (p ≤ 0.001) in case of diabetic group transplanted with IL-7 MSCs. Histological examination revealed enhanced vascular supply in skin tissues of diabetic animals transplanted with IL-7 transfected MSCs as compared to normal MSCs. Immunohistochemical results showed significantly higher expression of IL-7 (p ≤ 0.001) and α-smooth muscle actin(p ≤ 0.001) in the tissue sections of IL-7 transfected group as compared to normal MSCs and the diabetic control group; the latter indicates increase in the number of blood vessels. It is concluded from this study that IL-7 overexpression in MSCs can enhance the healing potential of MSCs and aid in wound closure in diabetic animals through the induction of angiogenic genes.
Collapse
Affiliation(s)
- Ramla Sana Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Midhat Batool Zaidi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nadia Naeem
- Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri, Suparco Road, KDA Scheme-33, Karachi, Pakistan
| | - Kanwal Haneef
- National Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| | - Rida-E-Maria Qazi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rakhshinda Habib
- Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri, Suparco Road, KDA Scheme-33, Karachi, Pakistan
| | - Tuba Shakil Malick
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Anwar Ali
- Department of Physiology, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
20
|
Sano E, Mori C, Nashimoto Y, Yokokawa R, Kotera H, Torisawa YS. Engineering of vascularized 3D cell constructs to model cellular interactions through a vascular network. BIOMICROFLUIDICS 2018; 12:042204. [PMID: 29861815 PMCID: PMC5955719 DOI: 10.1063/1.5027183] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/04/2018] [Indexed: 05/12/2023]
Abstract
Current in vitro 3D culture models lack a vascular system to transport oxygen and nutrients, as well as cells, which is essential to maintain cellular viability and functions. Here, we describe a microfluidic method to generate a perfusable vascular network that can form inside 3D multicellular spheroids and functionally connect to microchannels. Multicellular spheroids containing endothelial cells and lung fibroblasts were embedded within a hydrogel inside a microchannel, and then, endothelial cells were seeded into both sides of the hydrogel so that angiogenic sprouts from the cell spheroids and the microchannels were anastomosed to form a 3D vascular network. Solution containing cells and reagents can be perfused inside the cell spheroids through the vascular network by injecting it into a microchannel. This method can be used to study cancer cell migration towards 3D co-culture spheroids through a vascular network. We recapitulated a bone-like microenvironment by culturing multicellular spheroids containing osteo-differentiated mesenchymal stem cells (MSCs), as well as endothelial cells, and fibroblasts in the device. After the formation of vascularized spheroids, breast cancer cells were injected into a microchannel connected to a vascular network and cultured for 7 days on-chip to monitor cellular migration. We demonstrated that migration rates of the breast cancer cells towards multicellular spheroids via blood vessels were significantly higher in the bone-like microenvironment compared with the microenvironment formed by undifferentiated MSCs. These findings demonstrate the potential value of the 3D vascularized spheroids-on-a-chip for modeling in vivo-like cellular microenvironments, drug delivery through blood vessels, and cellular interactions through a vascular network.
Collapse
Affiliation(s)
- Emi Sano
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Chihiro Mori
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Yuji Nashimoto
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Hidetoshi Kotera
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Yu-suke Torisawa
- Author to whom correspondence should be addressed: . Tel.: +81-75-383-3701. Fax: +81-75-383-3681
| |
Collapse
|
21
|
Nashimoto Y, Teraoka Y, Banan Sadeghian R, Nakamasu A, Arima Y, Hanada S, Kotera H, Nishiyama K, Miura T, Yokokawa R. Perfusable Vascular Network with a Tissue Model in a Microfluidic Device. J Vis Exp 2018. [PMID: 29683439 DOI: 10.3791/57242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
A spheroid (a multicellular aggregate) is regarded as a good model of living tissues in the human body. Despite the significant advancement in the spheroid cultures, a perfusable vascular network in the spheroids remains a critical challenge for long-term culture required to maintain and develop their functions, such as protein expressions and morphogenesis. The protocol presents a novel method to integrate a perfusable vascular network within the spheroid in a microfluidic device. To induce a perfusable vascular network in the spheroid, angiogenic sprouts connected to microchannels were guided to the spheroid by utilizing angiogenic factors from human lung fibroblasts cultured in the spheroid. The angiogenic sprouts reached the spheroid, merged with the endothelial cells co-cultured in the spheroid, and formed a continuous vascular network. The vascular network could perfuse the interior of the spheroid without any leakage. The constructed vascular network may be further used as a route for supply of nutrients and removal of waste products, mimicking blood circulation in vivo. The method provides a new platform in spheroid culture toward better recapitulation of living tissues.
Collapse
Affiliation(s)
| | | | | | | | - Yuichiro Arima
- International Research Center for Medical Sciences (IRCMS), Kumamoto University
| | - Sanshiro Hanada
- International Research Center for Medical Sciences (IRCMS), Kumamoto University
| | | | - Koichi Nishiyama
- International Research Center for Medical Sciences (IRCMS), Kumamoto University
| | - Takashi Miura
- Graduate School of Medical Sciences, Kyushu University
| | | |
Collapse
|
22
|
Andrejecsk JW, Hughes CC. Engineering perfused microvascular networks into microphysiological systems platforms. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2018.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
23
|
Assessing angiogenic responses induced by primary human prostate stromal cells in a three-dimensional fibrin matrix assay. Oncotarget 2018; 7:71298-71308. [PMID: 27542256 PMCID: PMC5342079 DOI: 10.18632/oncotarget.11347] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/10/2016] [Indexed: 01/08/2023] Open
Abstract
Accurate modeling of angiogenesis in vitro is essential for guiding the preclinical development of novel anti-angiogenic agents and treatment strategies. The formation of new blood vessels is a multifactorial and multi-stage process dependent upon paracrine factors produced by stromal cells in the local microenvironment. Mesenchymal stem cells (MSCs) are multipotent cells in adults that can be recruited to sites of inflammation and tissue damage where they aid in wound healing through regenerative, trophic, and immunomodulatory properties. Primary stromal cultures derived from human bone marrow, normal prostate, or prostate cancer tissue are highly enriched in MSCs and stromal progenitors. Using conditioned media from these primary cultures, a robust pro-angiogenic response was observed in a physiologically-relevant three-dimensional fibrin matrix assay. To evaluate the utility of this assay, the allosteric HDAC4 inhibitor tasquinimod and the anti-VEGF monoclonal antibody bevacizumab were used as model compounds with distinct mechanisms of action. While both agents had a profound inhibitory effect on endothelial sprouting, only bevacizumab induced significant regression of established vessels. Additionally, the pro-angiogenic properties of MSCs derived from prostate cancer patients provides further evidence that selective targeting of this population may be of therapeutic benefit.
Collapse
|
24
|
Haque N, Abdullah BJJ, Kasim NHA. Secretome: Pharmaceuticals for Cell-Free Regenerative Therapy. STEM CELL DRUGS - A NEW GENERATION OF BIOPHARMACEUTICALS 2018. [DOI: 10.1007/978-3-319-99328-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Chung M, Ahn J, Son K, Kim S, Jeon NL. Biomimetic Model of Tumor Microenvironment on Microfluidic Platform. Adv Healthc Mater 2017; 6. [PMID: 28544639 DOI: 10.1002/adhm.201700196] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/03/2017] [Indexed: 12/18/2022]
Abstract
The "Tumor microenvironment" (TME) is a complex, interacting system of the tumor and its surrounding environment. The TME has drawn more attention recently in attempts to overcome current drug resistance and the recurrence of cancer by understanding the cancer and its microenvironment systematically, beyond past reductionist approaches. However, a lack of experimental tools to dissect the intricate interactions has hampered in-depth research into the TME. Here, a biomimetic TME model using a microfluidic platform is presented, which enables the interaction between TME constituents to be studied in a comprehensive manner. Paracrine interactions of cocultured tumor cell lines (SK-OV-3, MKN-74, and SW620) with primary fibroblasts show marked morphological changes in the tumor cells, depending on the type of tumor cells, and, importantly, the composition of the extracellular matrix. Furthermore, this model allows direct observation of angiogenesis induced by the tumor-stroma interaction. Finally, reconstituting simultaneous angiogenesis and lymphangiogenesis induced by the tumor-stromal interaction with TME mimicking extrinsic factors is enabled. It is believed that the in vitro biomimetic model and the experimental concepts described will help to shed light on the complex biology of the TME.
Collapse
Affiliation(s)
- Minhwan Chung
- Mechanical Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Jungho Ahn
- Mechanical Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Kyungmin Son
- Mechanical Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Sudong Kim
- Mechanical Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- Mechanical Engineering; Seoul National University; Seoul 08826 Republic of Korea
- Institute of Advanced Machines and Design; Seoul National University; Seoul 08826 Republic of Korea
| |
Collapse
|
26
|
Xu JG, Zhu SY, Heng BC, Dissanayaka WL, Zhang CF. TGF-β1-induced differentiation of SHED into functional smooth muscle cells. Stem Cell Res Ther 2017; 8:10. [PMID: 28114966 PMCID: PMC5260045 DOI: 10.1186/s13287-016-0459-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. Obtaining the appropriate building blocks for vascular tissue engineering (i.e. endothelial and mural cells) is a challenging task for tissue neovascularization. Hence, we investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs). METHODS We utilized two cytokines of the TGF-β family, transforming growth factor beta 1 (TGF-β1) and bone morphogenetic protein 4 (BMP4), to induce SHED differentiation into SMCs. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to assess mRNA expression, and protein expression was analyzed using flow cytometry, western blot and immunostaining. Additionally, to examine whether these SHED-derived SMCs possess the same function as primary SMCs, in vitro Matrigel angiogenesis assay, fibrin gel bead assay, and functional contraction study were used here. RESULTS By analyzing the expression of specific markers of SMCs (α-SMA, SM22α, Calponin, and SM-MHC), we confirmed that TGF-β1, and not BMP4, could induce SHED differentiation into SMCs. The differentiation efficiency was relatively high (α-SMA+ 86.1%, SM22α+ 93.9%, Calponin+ 56.8%, and SM-MHC+ 88.2%) as assessed by flow cytometry. In vitro Matrigel angiogenesis assay showed that the vascular structures generated by SHED-derived SMCs and human umbilical vein endothelial cells (HUVECs) were comparable to primary SMCs and HUVECs in terms of vessel stability. Fibrin gel bead assay showed that SHED-derived SMCs had a stronger capacity for promoting vessel formation compared with primary SMCs. Further analyses of protein expression in fibrin gel showed that cultures containing SHED-derived SMCs exhibited higher expression levels of Fibronectin than the primary SMCs group. Additionally, it was also confirmed that SHED-derived SMCs exhibited functional contractility. When SB-431542, a specific inhibitor of ALK5 was administered, TGF-β1 stimulation could not induce SHED into SMCs, indicating that the differentiation of SHED into SMCs is somehow related to the TGF-β1-ALK5 signaling pathway. CONCLUSIONS SHED could be successfully induced into functional SMCs for vascular tissue engineering, and this course could be regulated through the ALK5 signaling pathway. Hence, SHED appear to be a promising candidate cell type for vascular tissue engineering.
Collapse
Affiliation(s)
- Jian Guang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Shao Yue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Waruna Lakmal Dissanayaka
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Cheng Fei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| |
Collapse
|
27
|
Kim S, Chung M, Ahn J, Lee S, Jeon NL. Interstitial flow regulates the angiogenic response and phenotype of endothelial cells in a 3D culture model. LAB ON A CHIP 2016; 16:4189-4199. [PMID: 27722679 DOI: 10.1039/c6lc00910g] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
A crucial yet ill-defined phenomenon involved in the remodeling of vascular networks, including angiogenic sprouting, is flow-mediated endothelial dynamics and phenotype changes. Despite interstitial flow (IF) being ubiquitously present in living tissues surrounding blood capillaries, it is rarely investigated and poorly understood how endothelial cells respond to this flow during morphogenesis. Here we develop a microfluidic 3D in vitro model to investigate the role of IF during vasculogenic formation and angiogenic remodeling of microvascular networks. In the presented model, human blood endothelial cells co-cultured with stromal fibroblasts spontaneously organize into an interconnected microvascular network and then further expand to adjacent avascular regions in a manner of neovessel sprouting. We found that in the presence of IF, vasculogenic organization of the microvascular network was significantly facilitated regardless of the flow direction, whereas angiogenic sprouting was promoted only when the directions of flow and sprouting were opposite while angiogenic activity was suppressed into the direction of flow. We also observed that the vasculatures switch between active angiogenic remodeling and quiescent/non-sprouting state in the contexts provided by IF. This regulatory effect can be utilized to examine the role of anti-angiogenic compounds, clearly distinguishing the differential influences of the compounds depending on their mechanisms of action. Collectively, these results suggest that IF may serve as a critical regulator in tissue vascularization and pathological angiogenesis.
Collapse
Affiliation(s)
- Sudong Kim
- Mechanical Engineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea.
| | - Minhwan Chung
- Mechanical Engineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea.
| | - Jungho Ahn
- Mechanical Engineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea.
| | - Somin Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| | - Noo Li Jeon
- Mechanical Engineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea. and Interdisciplinary Program for Bioengineering, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| |
Collapse
|
28
|
Fujita H, Gomori A, Fujioka Y, Kataoka Y, Tanaka K, Hashimoto A, Suzuki T, Ito K, Haruma T, Yamamoto-Yokoi H, Harada N, Sakuragi M, Oda N, Matsuo K, Inada M, Yonekura K. High Potency VEGFRs/MET/FMS Triple Blockade by TAS-115 Concomitantly Suppresses Tumor Progression and Bone Destruction in Tumor-Induced Bone Disease Model with Lung Carcinoma Cells. PLoS One 2016; 11:e0164830. [PMID: 27736957 PMCID: PMC5063576 DOI: 10.1371/journal.pone.0164830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 10/01/2016] [Indexed: 01/25/2023] Open
Abstract
Approximately 25-40% of patients with lung cancer show bone metastasis. Bone modifying agents reduce skeletal-related events (SREs), but they do not significantly improve overall survival. Therefore, novel therapeutic approaches are urgently required. In this study, we investigated the anti-tumor effect of TAS-115, a VEGFRs and HGF receptor (MET)-targeted kinase inhibitor, in a tumor-induced bone disease model. A549-Luc-BM1 cells, an osteo-tropic clone of luciferase-transfected A549 human lung adenocarcinoma cells (A549-Luc), produced aggressive bone destruction associated with tumor progression after intra-tibial (IT) implantation into mice. TAS-115 significantly reduced IT tumor growth and bone destruction. Histopathological analysis showed a decrease in tumor vessels after TAS-115 treatment, which might be mediated through VEGFRs inhibition. Furthermore, the number of osteoclasts surrounding the tumor was decreased after TAS-115 treatment. In vitro studies demonstrated that TAS-115 inhibited HGF-, VEGF-, and macrophage-colony stimulating factor (M-CSF)-induced signaling pathways in osteoclasts. Moreover, TAS-115 inhibited Feline McDonough Sarcoma oncogene (FMS) kinase, as well as M-CSF and receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation. Thus, VEGFRs/MET/FMS-triple inhibition in osteoclasts might contribute to the potent efficacy of TAS-115. The fact that concomitant dosing of sunitinib (VEGFRs/FMS inhibition) with crizotinib (MET inhibition) exerted comparable inhibitory efficacy for bone destruction to TAS-115 also supports this notion. In conclusion, TAS-115 inhibited tumor growth via VEGFR-kinase blockade, and also suppressed bone destruction possibly through VEGFRs/MET/FMS-kinase inhibition, which resulted in potent efficacy of TAS-115 in an A549-Luc-BM1 bone disease model. Thus, TAS-115 shows promise as a novel therapy for lung cancer patients with bone metastasis.
Collapse
Affiliation(s)
- Hidenori Fujita
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
- * E-mail:
| | - Akira Gomori
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yayoi Fujioka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yuki Kataoka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kenji Tanaka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Akihiro Hashimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Takamasa Suzuki
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kenjiro Ito
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Tomonori Haruma
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hiromi Yamamoto-Yokoi
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Naomoto Harada
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Motomu Sakuragi
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Nobuyuki Oda
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kenichi Matsuo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Masaki Inada
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
| | - Kazuhiko Yonekura
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| |
Collapse
|
29
|
Arulmoli J, Wright HJ, Phan DTT, Sheth U, Que RA, Botten GA, Keating M, Botvinick EL, Pathak MM, Zarembinski TI, Yanni DS, Razorenova OV, Hughes CCW, Flanagan LA. Combination scaffolds of salmon fibrin, hyaluronic acid, and laminin for human neural stem cell and vascular tissue engineering. Acta Biomater 2016; 43:122-138. [PMID: 27475528 PMCID: PMC5386322 DOI: 10.1016/j.actbio.2016.07.043] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVβ1 and α5β1, and several laminin binding integrins (α7β1, α6β1, α3β1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.
Collapse
Affiliation(s)
- Janahan Arulmoli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Heather J Wright
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Duc T T Phan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Urmi Sheth
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard A Que
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Giovanni A Botten
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark Keating
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Medha M Pathak
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Daniel S Yanni
- Disc Comfort, Inc., 351 Hospital Road, Suite 202, Newport Beach, CA 92663, USA
| | - Olga V Razorenova
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lisa A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
30
|
Ziegler ME, Hatch MMS, Wu N, Muawad SA, Hughes CCW. mTORC2 mediates CXCL12-induced angiogenesis. Angiogenesis 2016; 19:359-71. [PMID: 27106789 DOI: 10.1007/s10456-016-9509-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/03/2016] [Indexed: 01/26/2023]
Abstract
The chemokine CXCL12, through its receptor CXCR4, positively regulates angiogenesis by promoting endothelial cell (EC) migration and tube formation. However, the relevant downstream signaling pathways in EC have not been defined. Similarly, the upstream activators of mTORC2 signaling in EC are also poorly defined. Here, we demonstrate for the first time that CXCL12 regulation of angiogenesis requires mTORC2 but not mTORC1. We find that CXCR4 signaling activates mTORC2 as indicated by phosphorylation of serine 473 on Akt and does so through a G-protein- and PI3K-dependent pathway. Significantly, independent disruption of the mTOR complexes by drugs or multiple independent siRNAs reveals that mTORC2, but not mTORC1, is required for microvascular sprouting in a 3D in vitro angiogenesis model. Importantly, in a mouse model, both tumor angiogenesis and tumor volume are significantly reduced only when mTORC2 is inhibited. Finally, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), which is a key regulator of glycolytic flux, is required for microvascular sprouting in vitro, and its expression is reduced in vivo when mTORC2 is targeted. Taken together, these findings identify mTORC2 as a critical signaling nexus downstream of CXCL12/CXCR4 that represents a potential link between mTORC2, metabolic regulation, and angiogenesis.
Collapse
Affiliation(s)
- Mary E Ziegler
- The Department of Molecular Biology and Biochemistry, University of California Irvine, 3219 McGaugh Hall, Mail Code: 3900, Irvine, CA, 92697, USA
| | - Michaela M S Hatch
- The Department of Molecular Biology and Biochemistry, University of California Irvine, 3219 McGaugh Hall, Mail Code: 3900, Irvine, CA, 92697, USA
| | - Nan Wu
- The Department of Molecular Biology and Biochemistry, University of California Irvine, 3219 McGaugh Hall, Mail Code: 3900, Irvine, CA, 92697, USA
| | - Steven A Muawad
- The Department of Molecular Biology and Biochemistry, University of California Irvine, 3219 McGaugh Hall, Mail Code: 3900, Irvine, CA, 92697, USA
| | - Christopher C W Hughes
- The Department of Molecular Biology and Biochemistry, University of California Irvine, 3219 McGaugh Hall, Mail Code: 3900, Irvine, CA, 92697, USA. .,The Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, USA. .,The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
31
|
Stromal vascular fraction: A regenerative reality? Part 2: Mechanisms of regenerative action. J Plast Reconstr Aesthet Surg 2015; 69:180-8. [PMID: 26546112 DOI: 10.1016/j.bjps.2015.10.014] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/26/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022]
Abstract
Adipose tissue is a rich source of cells with emerging promise for tissue engineering and regenerative medicine. The stromal vascular fraction (SVF), in particular, is an eclectic composite of cells with progenitor activity that includes preadipocytes, mesenchymal stem cells, pericytes, endothelial cells, and macrophages. SVF has enormous potential for therapeutic application and is being investigated for multiple clinical indications including lipotransfer, diabetes-related complications, nerve regeneration, burn wounds and numerous others. In Part 2 of our review, we explore the basic science behind the regenerative success of the SVF and discuss significant mechanisms that are at play. The existing literature suggests that angiogenesis, immunomodulation, differentiation, and extracellular matrix secretion are the main avenues through which regeneration and healing is achieved by the stromal vascular fraction.
Collapse
|
32
|
Chang F, Lemmon CA, Nilaratanakul V, Rotter V, Romer L. Endothelial matrix assembly during capillary morphogenesis: insights from chimeric TagRFP-fibronectin matrix. J Histochem Cytochem 2014; 62:774-90. [PMID: 25063001 PMCID: PMC4209295 DOI: 10.1369/0022155414547419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/13/2014] [Indexed: 11/22/2022] Open
Abstract
Biologically relevant, three-dimensional extracellular matrix is an essential component of in vitro vasculogenesis models. WI-38 fibroblasts assemble a 3D matrix that induces endothelial tubulogenesis, but this model is challenged by fibroblast senescence and the inability to distinguish endothelial cell-derived matrix from matrix made by WI-38 fibroblasts. Matrices produced by hTERT-immortalized WI-38 recapitulated those produced by wild type fibroblasts. ECM fibrils were heavily populated by tenascin-C, fibronectin, and type VI collagen. Nearly half of the total type I collagen, but only a small fraction of the type IV collagen, were incorporated into ECM. Stable hTERT-WI-38 transfectants expressing TagRFP-fibronectin incorporated TagRFP into ~90% of the fibronectin in 3D matrices. TagRFP-fibronectin colocalized with tenascin-C and with type I collagen in a pattern that was similar to that seen in matrices from wild type WI-38. Human Umbilical Vein Endothelial Cells (HUVEC) formed 3D adhesions and tubes on WI38-hTERT-TagRFP-FN-derived matrices, and the TagRFP-fibronectin component of this new 3D human fibroblast matrix model facilitated the demonstration of concentrated membrane type 1 metalloprotease and new HUVEC FN and collagen type IV fibrils during EC tubulogenesis. These findings indicate that WI-38-hTERT- and WI-38-hTERT-TagRFP-FN-derived matrices provide platforms for the definition of new matrix assembly and remodeling events during vasculogenesis.
Collapse
Affiliation(s)
- Fumin Chang
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Christopher A Lemmon
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Voraphoj Nilaratanakul
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Varda Rotter
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Lewis Romer
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| |
Collapse
|
33
|
Lim SH, Kim C, Aref AR, Kamm RD, Raghunath M. Complementary effects of ciclopirox olamine, a prolyl hydroxylase inhibitor and sphingosine 1-phosphate on fibroblasts and endothelial cells in driving capillary sprouting. Integr Biol (Camb) 2014; 5:1474-84. [PMID: 24190477 DOI: 10.1039/c3ib40082d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Capillary sprouting, a key step of neoangiogenesis in wound healing and tumor growth, also represents a therapeutic target for tissue repair. It requires crosstalk between endothelial cells (EC) and other cell types. We studied this process in a microfluidic platform that allows EC to migrate out of a channel across a collagen gel up a gradient of factors produced by a collection of encapsulated fibroblasts. Introduction of a prolyl hydroxylase inhibitor (PHi), ciclopirox olamine (CPX) to stabilize hypoxia inducible factor 1α (HIF-1α) predominantly in fibroblasts induced capillary sprouting in EC, but the most complex tubular networks with true lumina formed after combining CPX with the lysophospholipid sphingosine 1-phosphate (S1P). The enhanced angiogenesis is a possible consequence of the generation of mutually stimulating factors as each cell type responded differently to the compounds. The combination of CPX and S1P induced secretion of vascular endothelial growth factor (VEGF) in fibroblast culture whereas the angiogenic monocyte chemoattractant protein (MCP)-1 was exclusively secreted by fibroblasts, but only in the presence of EC-conditioned medium. Antibody interference with fibroblast-produced VEGF and MCP-1 inhibited the sprouting response. These observations not only demonstrate the collaboration of EC and fibroblasts in inducing capillary sprouting but also suggest that the combination of CPX and S1P enhances angiogenesis and thus might be of therapeutic value for the pharmacological induction of tissue repair and regeneration.
Collapse
Affiliation(s)
- Sei Hien Lim
- Biosystems & Micromechanics Interdisciplinary Research Group (BioSyM), Singapore-MIT Alliance in Research & Technology (SMART), Singapore
| | | | | | | | | |
Collapse
|
34
|
Ehsan SM, Welch-Reardon KM, Waterman ML, Hughes CCW, George SC. A three-dimensional in vitro model of tumor cell intravasation. Integr Biol (Camb) 2014; 6:603-10. [PMID: 24763498 PMCID: PMC4046910 DOI: 10.1039/c3ib40170g] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metastasis is the cause of over 90% of all human cancer deaths. Early steps in the metastatic process include: the formation of new blood vessels, the initiation of epithelial-mesenchymal transition (EMT), and the mobilization of tumor cells into the circulation. There are ongoing efforts to replicate the physiological landscape of human tumor tissue using three-dimensional in vitro culture models; however, few systems are able to capture the full range of authentic, complex in vivo events such as neovascularization and intravasation. Here we introduce the Prevascularized Tumor (PVT) model to investigate early events of solid tumor progression. PVT spheroids are composed of endothelial and tumor cells, and are embedded in a fibrin matrix containing fibroblasts. The PVT model facilitates two mechanisms of vessel formation: robust sprouting angiogenesis into the matrix, and contiguous vascularization within the spheroid. Furthermore, the PVT model enables the intravasation of tumor cells that is enhanced under low oxygen conditions and is also dependent on the key EMT transcription factor Slug. The PVT model provides a significant advance in the mimicry of human tumors in vitro, and may improve investigation and targeting of events in the metastatic process.
Collapse
Affiliation(s)
- Seema M Ehsan
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|
35
|
Yang X, Zhang XF, Lu X, Jia HL, Liang L, Dong QZ, Ye QH, Qin LX. MicroRNA-26a suppresses angiogenesis in human hepatocellular carcinoma by targeting hepatocyte growth factor-cMet pathway. Hepatology 2014; 59:1874-1885. [PMID: 24259426 DOI: 10.1002/hep.26941] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/17/2013] [Indexed: 12/16/2022]
Abstract
UNLABELLED MicroRNA (miR)-26a can suppress tumor growth and metastasis of hepatocellular carcinoma (HCC). Since angiogenesis is important for tumor growth and metastasis, we investigated the possible roles of miR-26a in tumor angiogenesis. Down-regulation of miR-26a was found to correlate with an increased angiogenic potential of HCC. Through gain- and loss-of-function studies, miR-26a was demonstrated to significantly inhibit vascular endothelial growth factor A (VEGFA) expression in HCC cells and then suppress the promoting effects of HCC cells on in vitro proliferation, migration, and capillary tube formation of endothelial cells, as well as in vivo tumor angiogenesis of HCC. Hepatocyte growth factor (HGF) was identified as a target of miR-26a. HGF simulation antagonized the effects induced by miR-26a up-regulation. In contrast, silencing HGF induced similar effects to miR-26a. We further found that miR-26a exerted its antiangiogenesis function, at least in part, by inhibiting HGF-hepatocyte growth factor receptor (cMet) and its downstream signaling pathway, in turn, suppressing VEGFA production in HCC cells and impairing VEGFR2-signaling in endothelial cells. HCC patients who had high miR-26a, low HGF, low VEGFA, or low microvessel density (MVD) in tumor tissues had a better prognosis with longer overall survival (OS) and time to recurrence (TTR). In multivariate analysis, miR-26a, or in combination with HGF, was demonstrated to be an independent prognostic indicator for OS and TTR of HCC patients. CONCLUSION miR-26a could suppress tumor angiogenesis of HCC through HGF-cMet signaling, and it is a new hopeful therapeutic target and prognostic marker for HCC.
Collapse
Affiliation(s)
- Xin Yang
- Liver Cancer Institute & Zhongshan Hospital, Institutes of Biomedical Science, Fudan University, Shanghai, China. Key Laboratory of Carcinogenesis & Cancer Invasion, Ministry of Education, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
There is significant interest within the tissue engineering and pharmaceutical industries to create 3D microphysiological systems of human organ function. The interest stems from a growing concern that animal models and simple 2D culture systems cannot replicate essential features of human physiology that are critical to predict drug response, or simply to develop new therapeutic strategies to repair or replace damaged organs. Central to human organ function is a microcirculation that not only enhances the rate of nutrient and waste transport by convection, but also provides essential additional physiological functions that can be specific to each organ. This review highlights progress in the creation of in vitro functional microvessel networks, and emphasizes organ-specific functional and structural characteristics that should be considered in the future mimicry of four organ systems that are of primary interest: lung, brain, liver, and muscle (skeletal and cardiac).
Collapse
|
37
|
Mauge L, Sabatier F, Boutouyrie P, D'Audigier C, Peyrard S, Bozec E, Blanchard A, Azizi M, Dizier B, Dignat-George F, Gaussem P, Smadja DM. Forearm ischemia decreases endothelial colony-forming cell angiogenic potential. Cytotherapy 2014; 16:213-24. [DOI: 10.1016/j.jcyt.2013.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 09/18/2013] [Accepted: 09/28/2013] [Indexed: 01/03/2023]
|
38
|
Locher R, Erba PA, Hirsch B, Bombardieri E, Giovannoni L, Neri D, Dürkop H, Menssen HD. Abundant in vitro expression of the oncofetal ED-B-containing fibronectin translates into selective pharmacodelivery of (131)I-L19SIP in a prostate cancer patient. J Cancer Res Clin Oncol 2014; 140:35-43. [PMID: 24132461 DOI: 10.1007/s00432-013-1538-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 10/07/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE The extradomain B of fibronectin (ED-B) is a promising vascular target for selective pharmacodelivery in cancer patients. We analyzed a large series of prostatectomies from patients with prostate cancer, hyperplastic prostate disease, and normal prostates to study extent and tumor-selectivity of ED-B expression. METHODS Using immunohistology, 68 adenocarcinomas of the prostate or prostate cancer-inflicted lymph nodes, 4 samples of benign prostatic hyperplasia, and 6 normal prostate glands were studied for ED-B expressing newly formed blood vessels. Further, we treated an advanced prostate cancer patient with the anti-ED-B antibody (131)I-L19SIP to study in vivo target accessibility. RESULTS ED-B-positive blood vessels were found significantly more frequent in prostate cancers as compared with peritumoral prostate tissues or normal prostate glands, independent of tumor differentiation. The ED-B-positive blood vessels' density was 97 (±23), 65 (±9), and 59 (±9)/mm(2) in G3, G2, and G1 prostate cancers, respectively, and 7 (±5)/mm(2) in normal prostate glands. In high-grade (G3) prostate cancers, also the peritumoral tissue showed a higher density of ED-B vessels than normal prostate glands. Similar results were obtained when ED-B-positive vessel density was expressed as a fraction of CD34-positive vessel density. Finally, selective uptake of ED-B-binding (131)I-L19SIP to tumor lesions was found in an advanced prostate cancer patient by whole-body planar scintigraphy. CONCLUSIONS ED-B-positive blood vessels were found to a large extent in prostate cancer tissues, but only rarely in normal prostates or benign prostatic hyperplasia. Whole-body planar scintigraphy in a prostate cancer patient confirmed selective uptake of (131)I-L19SIP in the prostate cancer tissues, qualifying ED-B as a promising target for selective pharmacodelivery of anticancer agents in prostate cancer.
Collapse
|
39
|
Reyes-Botero G, Dehais C, Idbaih A, Martin-Duverneuil N, Lahutte M, Carpentier C, Letouzé E, Chinot O, Loiseau H, Honnorat J, Ramirez C, Moyal E, Figarella-Branger D, Ducray F. Contrast enhancement in 1p/19q-codeleted anaplastic oligodendrogliomas is associated with 9p loss, genomic instability, and angiogenic gene expression. Neuro Oncol 2013; 16:662-70. [PMID: 24353325 DOI: 10.1093/neuonc/not235] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The aim of this study was to correlate MRI features and molecular characteristics in anaplastic oligodendrogliomas (AOs). METHODS The MRI characteristics of 50 AO patients enrolled in the French national network for high-grade oligodendroglial tumors were analyzed. The genomic profiles and IDH mutational statuses were assessed using high-resolution single-nucleotide polymorphism arrays and direct sequencing, respectively. The gene expression profiles of 25 1p/19q-codeleted AOs were studied on Affymetrix expression arrays. RESULTS Most of the cases were frontal lobe contrast-enhanced tumors (52%), but the radiological presentations of these cases were heterogeneous, ranging from low-grade glioma-like aspects (26%) to glioblastoma-like aspects (22%). The 1p/19q codeletion (n = 39) was associated with locations in the frontal lobe (P = .001), with heterogeneous intratumoral signal intensities (P = .003) and with no or nonmeasurable contrast enhancements (P = .01). The IDH wild-type AOs (n = 7) more frequently displayed ringlike contrast enhancements (P = .03) and were more frequently located outside of the frontal lobe (P = .01). However, no specific imaging pattern could be identified for the 1p/19q-codeleted AO or the IDH-mutated AO. Within the 1p/19q-codeleted AO, the contrast enhancement was associated with larger tumor volumes (P = .001), chromosome 9p loss and CDKN2A loss (P = .006), genomic instability (P = .03), and angiogenesis-related gene expression (P < .001), particularly for vascular endothelial growth factor A and angiopoietin 2. CONCLUSION In AOs, the 1p/19q codeletion and the IDH mutation are associated with preferential (but not with specific) imaging characteristics. Within 1p/19q-codeleted AO, imaging heterogeneity is related to additional molecular alterations, especially chromosome 9p loss, which is associated with contrast enhancement and larger tumor volume.
Collapse
Affiliation(s)
- German Reyes-Botero
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France (G.R.B., C.D., A.I.); Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epinière, Paris, France (A.I., C.C.); AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neuro-radiologie, Paris, France (N.M.-D.); Service de Santé des Armées, Hôpital d'Instruction des Armées, Paris, France (M.L.); Programme Carte d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France (E.L.); AP-HM, Hôpital de la Timone, Service de Neuro-Oncologie, Marseille , France (O.C.); CHU Bordeaux, Hôpital Pellegrin, Service de Neurochirurgie, Bordeaux, France (H.L.); Hospices Civils de Lyon, Hôpital Pierre Wertheimer, Service de Neuro-Oncologie, Bron, France (J.H., F.D.); INSERM U1028, CNRS UMR5292, Bron, France (J.H., F.D.); CHU Lille, Hôpital Roger Salengro, Clinique de Neurochirurgie, Lille, France (C.R.); Institut Claudius Regaud, Département de Radiothérapie, Toulouse, France (E.M.); AP-HM, Hôpital de la Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France (D.F.-B.); Université de la Méditerranée, Aix-Marseille, Faculté de Médecine La Timone, Marseille, France (D.F.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Harvey A, Yen TY, Aizman I, Tate C, Case C. Proteomic analysis of the extracellular matrix produced by mesenchymal stromal cells: implications for cell therapy mechanism. PLoS One 2013; 8:e79283. [PMID: 24244468 PMCID: PMC3828366 DOI: 10.1371/journal.pone.0079283] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/27/2013] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) transiently transfected with notch1 intracellular domain (NICD) are beneficial for neurological disorders as observed in several preclinical studies. Extracellular matrix (ECM) derived from NICD-transfected MSCs has been previously shown to support in vitro neural cell growth and survival better than that of un-transfected MSCs. To understand the underlying mechanism(s) by which NICD-transfected MSC-derived ECM supports neural cell growth and survival, we investigated the differences in NICD-transfected MSC- and MSC-derived ECM protein quantity and composition. To compare the ECM derived from MSCs and NICD-transfected MSCs, the proteins were sequentially solubilized using sodium dodecyl sulfate (SDS) and urea, quantified, and compared across four human donors. We then analyzed ECM proteins using either in-gel digests or in-solution surfactant-assisted trypsin digests (SAISD) coupled with reverse phase nano-liquid chromatography and tandem mass spectrometry (nLC-MS/MS). Analyses using nLC-MS/MS identified key components of ECM from NICD-transfected MSCs and un-transfected MSCs and revealed significant differences in their respective compositions. This work provides a reproducible method for identifying and comparing in vitro cell-derived ECM proteins, which is crucial for exploring the mechanisms underlying cellular therapy.
Collapse
Affiliation(s)
- Adam Harvey
- SanBio Inc., Mountain View, California, United States of America
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Ten-Yang Yen
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California, United States of America
| | - Irina Aizman
- SanBio Inc., Mountain View, California, United States of America
| | - Ciara Tate
- SanBio Inc., Mountain View, California, United States of America
| | - Casey Case
- SanBio Inc., Mountain View, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Norrby K. Metronomic chemotherapy and anti-angiogenesis: can upgraded pre-clinical assays improve clinical trials aimed at controlling tumor growth? APMIS 2013; 122:565-79. [PMID: 24164171 PMCID: PMC4282375 DOI: 10.1111/apm.12201] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/06/2013] [Indexed: 12/21/2022]
Abstract
Metronomic chemotherapy, which is continuously administered systemically at close to non-toxic doses, targets the endothelial cells (ECs) that are proliferating during tumor angiogenesis. This leads to harmful effects of an even greatly increased number contiguous tumor cells. Although pre-clinical studies of angiogenesis-related EC features in vitro and of the anti-angiogenic and anti-tumor effects in vivo of metronomic chemotherapy have provided valuable insights, clinical trials with this type of therapy have been less successful in inhibiting tumor growth. One possible reason for the apparent disconnect between the pre-clinical and clinical outcomes is that most of the currently used experimental angiogenesis assays and tumor models are incapable of yielding data that can be translated readily into the clinical setting. Many of the assays used suffer from unintentional artifactual effects, e.g., oxidative stress in vitro, and inflammation in vivo, which reduces the sensitivity and discriminatory power of the assays. Co-treatment with an antioxidant or the inclusion of antioxidants in the vehicle often significantly affects the angiogenesis-modulating outcome of metronomic mono-chemotherapy in vivo. This ‘metronomic chemotherapy vehicle factor’ merits further study, as do the observations of antagonistic effects following metronomic treatment with a combination of standard chemotherapeutic drugs in vivo.
Collapse
Affiliation(s)
- Klas Norrby
- Department of Pathology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
42
|
Chang WG, Andrejecsk JW, Kluger MS, Saltzman WM, Pober JS. Pericytes modulate endothelial sprouting. Cardiovasc Res 2013; 100:492-500. [PMID: 24042014 DOI: 10.1093/cvr/cvt215] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIM Angiogenic sprouts arise from microvessels formed by endothelial cells (ECs) invested by pericytes (PCs). The aim of this study was to examine the role of PCs in angiogenic sprouting, an understudied phenomenon. METHODS AND RESULTS We adapted a human EC spheroid model to examine PC effects on vascular endothelial growth factor-A-induced EC sprouting in vitro by using Bcl-2-transduced human umbilical vein ECs to reduce apoptosis in collagen gels. Human placental PCs, separated from endothelial spheroids by a transwell, or addition of PC-conditioned media increased EC sprouting primarily through hepatocyte growth factor (HGF). Mixed endothelial-PC spheroids formed similar numbers of endothelial sprouts as endothelial spheroids but the sprouts from mixed spheroids were invested by PCs within 24 h. PCs were recruited to the sprouts by platelet-derived growth factor (PDGF)-BB; inhibition of PDGF signalling reduced PC coverage and increased EC sprouting. Transplanted endothelial spheroids give rise to sprouts in vivo that evolve into perfused microvessels. Mixed endothelial-PC spheroids form similar numbers of microvessels as endothelial-only spheroids, but acquire human PC investment and have reduced average lumen diameter. CONCLUSIONS PCs promote endothelial sprouting by elaborating HGF, but when recruited to invest endothelial sprouts by PDGF-BB, limit the extent of sprouting in vitro and lumen diameter in vivo.
Collapse
Affiliation(s)
- William G Chang
- Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|