1
|
Buranawat B, Shaalan A, Garna DF, Di Silvio L. Development of Prevascularized Synthetic Block Graft for Maxillofacial Reconstruction. J Funct Biomater 2025; 16:18. [PMID: 39852574 PMCID: PMC11766148 DOI: 10.3390/jfb16010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Cranio-maxillofacial bone reconstruction, especially for large defects, remains challenging. Synthetic biomimetic materials are emerging as alternatives to autogenous grafts. Tissue engineering aims to create natural tissue-mimicking materials, with calcium phosphate-based scaffolds showing promise for bone regeneration applications. This study developed a porous calcium metaphosphate (CMP) scaffold with physicochemical properties mimicking natural bone, aiming to create a prevascularized synthetic bone graft. The scaffold, fabricated using sintered monocalcium phosphate with poly (vinyl alcohol) as a porogen, exhibited pore sizes ranging from 0 to 400 μm, with the highest frequency between 80 and 100 μm. The co-culture of endothelial cells (ECs) with human alveolar osteoblasts (aHOBs) on the scaffold led to the formation of tube-like structures and intrinsic VEGF release, reaching 10,455.6 pg/mL This level approached the optimal dose for vascular formation. Conversely, the co-culture with mesenchymal stem cells did not yield similar results. Combining ECs and aHOBs in the CMP scaffold offers a promising approach to developing prevascularized grafts for cranio-maxillofacial reconstruction. This innovative strategy can potentially enhance vascularization in large tissue-engineered constructs, addressing a critical limitation in current bone regeneration techniques. The prevascularized synthetic bone graft developed in this study could significantly improve the success rate of maxillofacial reconstructions, offering a viable alternative to autogenous grafts.
Collapse
Affiliation(s)
- Borvornwut Buranawat
- Center for Implant Dentistry and Periodontics, Faculty of Dentistry and Research Unit in Innovations in Periodontics, Oral Surgery and Advanced Technology in Implant Dentistry, Thammasat University, Bangkok 10200, Thailand;
| | - Abeer Shaalan
- Center for Oral, Clinical and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.S.); (D.F.G.)
| | - Devy F. Garna
- Center for Oral, Clinical and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.S.); (D.F.G.)
| | - Lucy Di Silvio
- Center for Oral, Clinical and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.S.); (D.F.G.)
| |
Collapse
|
2
|
Lyyra I, Isomäki M, Huhtala H, Kellomäki M, Miettinen S, Massera J, Sartoneva R. Ionic Dissolution Products of Lithium-, Strontium-, and Boron-Substituted Silicate Glasses Influence the Viability and Proliferation of Adipose Stromal Cells, Fibroblasts, Urothelial and Endothelial Cells. ACS OMEGA 2024; 9:49348-49367. [PMID: 39713681 PMCID: PMC11656255 DOI: 10.1021/acsomega.4c06587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/11/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024]
Abstract
While bioactive glasses (BaGs) have been studied mainly for bone applications, studies have also shown their potential for soft tissue engineering. Incorporating therapeutic ions, such as lithium (Li+), strontium (Sr2+), and boron (B3+) into the BaGs, has been found to promote angiogenesis and wound healing. However, a systematic study on the impact of Li+, Sr2+, B3+, and the other ions in the BaGs, has not been conducted on a wide range of cells. Although the interactions between the BaGs and cells have been studied, it is difficult to compare the results between studies and conclude the impact of BaGs between cell types due to the variability of culture conditions, cells, and materials. We aim to evaluate the dissolution behavior of Li-, Sr-, and B-substituted BaGs and the effects of their ionic dissolution products on the viability, proliferation, and morphology of multiple cell types: human adipose stromal cells (hASCs), human lung fibroblasts (cell line WI-38), human urothelial cells (hUCs), and human umbilical vein endothelial cells (HUVECs). In the dissolution study, the B-substituted glasses induced a higher increase in pH and released more ions than the silicate glasses. The undiluted BaG extracts supported the viability and proliferation of all the other cell types except the hUCs. Diluting the BaG extracts to 1:10 restored the viability of hUCs but induced distinctive morphological changes. Diluting the extracts more (1:100) almost fully restored the hUC morphology. To conclude, the ionic dissolution products of Li-, Sr-, and B-substituted BaGs seem beneficial for hASCs, WI-38, hUCs, and HUVECs, but attention must be paid to the ion concentrations.
Collapse
Affiliation(s)
- Inari Lyyra
- Faculty of
Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 3, Tampere FI-33720, Finland
| | - Mari Isomäki
- Faculty of
Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 3, Tampere FI-33720, Finland
| | - Heini Huhtala
- Faculty of
Social Sciences, Tampere University, Arvo Ylpön katu 34, Tampere FI-33520, Finland
| | - Minna Kellomäki
- Faculty of
Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 3, Tampere FI-33720, Finland
| | - Susanna Miettinen
- Faculty of
Medicine and Health Technology, Tampere
University, Arvo Ylpön katu 34, Tampere FI-33520, Finland
- Research
and Development and Innovation, Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Arvo Ylpön katu 6, Tampere FI-33521, Finland
| | - Jonathan Massera
- Faculty of
Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 3, Tampere FI-33720, Finland
| | - Reetta Sartoneva
- Faculty of
Medicine and Health Technology, Tampere
University, Arvo Ylpön katu 34, Tampere FI-33520, Finland
- Research
and Development and Innovation, Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Arvo Ylpön katu 6, Tampere FI-33521, Finland
- Department
of Obstetrics and Gynaecology, Seinäjoki Central Hospital, South Ostrobothnia Wellbeing Services County, Hanneksenrinne 7, Seinäjoki FI-60220, Finland
| |
Collapse
|
3
|
Crouch EE, Diafos LN, Valenzuela EJ, Wedderburn-Pugh K, Birrueta JO, Caston J, Joseph T, Andrews MG, Bhaduri A, Huang EJ. Profiling human brain vascular cells using single-cell transcriptomics and organoids. Nat Protoc 2024; 19:603-628. [PMID: 38102365 PMCID: PMC11537086 DOI: 10.1038/s41596-023-00929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/13/2023] [Indexed: 12/17/2023]
Abstract
Angiogenesis and neurogenesis are functionally interconnected during brain development. However, the study of the vasculature has trailed other brain cell types because they are delicate and of low abundance. Here we describe a protocol extension to purify prenatal human brain endothelial and mural cells with FACS and utilize them in downstream applications, including transcriptomics, culture and organoid transplantation. This approach is simple, efficient and generates high yields from small amounts of tissue. When the experiment is completed within a 24 h postmortem interval, these healthy cells produce high-quality data in single-cell transcriptomics experiments. These vascular cells can be cultured, passaged and expanded for many in vitro assays, including Matrigel vascular tube formation, microfluidic chambers and metabolic measurements. Under these culture conditions, primary vascular cells maintain expression of cell-type markers for at least 3 weeks. Finally, we describe how to use primary vascular cells for transplantation into cortical organoids, which captures key features of neurovascular interactions in prenatal human brain development. In terms of timing, tissue processing and staining requires ~3 h, followed by an additional 3 h of FACS. The transplant procedure of primary, FACS-purified vascular cells into cortical organoids requires an additional 2 h. The time required for different transcriptomic and epigenomic protocols can vary based on the specific application, and we offer strategies to mitigate batch effects and optimize data quality. In sum, this vasculo-centric approach offers an integrated platform to interrogate neurovascular interactions and human brain vascular development.
Collapse
Affiliation(s)
- Elizabeth E Crouch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Biomedical Science Graduate Program, University of California San Francisco, San Francisco, CA, USA.
| | - Loukas N Diafos
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Edward J Valenzuela
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Kaylee Wedderburn-Pugh
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Biomedical Science Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | - Janeth Ochoa Birrueta
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Jaela Caston
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Biomedical Science Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Tara Joseph
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Madeline G Andrews
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Aparna Bhaduri
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Eric J Huang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Biomedical Science Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
- Pathology Service 113B, San Francisco Veterans Affairs Healthcare System, San Francisco, CA, USA.
| |
Collapse
|
4
|
Hautanen V, Toimela T, Paparella M, Heinonen T. A Human Cell-based Assay to Assess the Induction of Vasculature Formation for Non-genotoxic Carcinogenicity Testing Purposes: A Pilot Study. Altern Lab Anim 2023:2611929231171165. [PMID: 37125451 DOI: 10.1177/02611929231171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The induction of vasculature formation is proposed to be a significant mechanism behind the non-genotoxic carcinogenicity of a chemical. The vasculature formation model used in this study is based on the coculture of human primary HUVECs and hASCs. This model was used to develop an assay to assess the induction of vasculature formation. Three assay protocols, based on different conditions, were developed and compared in order to identify the optimal conditions required. Some serum supplements and growth factors were observed to be essential for initiating vasculature formation. Of the studied putative positive reference chemicals, aspartame, sodium nitrite, bisphenol A and nicotine treatment led to a clear induction of vasculature formation, but arsenic and cadmium treatment only led to a slight increase. This human cell-based assay has the potential to be used as one test within a next generation testing battery, to assess the non-genotoxic carcinogenicity of a chemical through the mechanism of vasculature formation induction.
Collapse
Affiliation(s)
- Veera Hautanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tarja Toimela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Tuula Heinonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
5
|
Truelsen SLB, Mousavi N, Wei H, Harvey L, Stausholm R, Spillum E, Hagel G, Qvortrup K, Thastrup O, Harling H, Mellor H, Thastrup J. The cancer angiogenesis co-culture assay: In vitro quantification of the angiogenic potential of tumoroids. PLoS One 2021; 16:e0253258. [PMID: 34234354 PMCID: PMC8263287 DOI: 10.1371/journal.pone.0253258] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/01/2021] [Indexed: 01/07/2023] Open
Abstract
The treatment response to anti-angiogenic agents varies among cancer patients and predictive biomarkers are needed to identify patients with resistant cancer or guide the choice of anti-angiogenic treatment. We present “the Cancer Angiogenesis Co-Culture (CACC) assay”, an in vitro Functional Precision Medicine assay which enables the study of tumouroid induced angiogenesis. This assay can quantify the ability of a patient-derived tumouroid to induce vascularization by measuring the induction of tube formation in a co-culture of vascular cells and tumoroids established from the primary colorectal tumour or a metastasis. Furthermore, the assay can quantify the sensitivity of patient-derived tumoroids to anti-angiogenic therapies. We observed that tube formation increased in a dose-dependent manner upon treatment with the pro-angiogenic factor vascular endothelial growth factor A (VEGF-A). When investigating the angiogenic potential of tumoroids from 12 patients we found that 9 tumoroid cultures induced a significant increase in tube formation compared to controls without tumoroids. In these 9 angiogenic tumoroid cultures the tube formation could be abolished by treatment with one or more of the investigated anti-angiogenic agents. The 3 non-angiogenic tumoroid cultures secreted VEGF-A but we observed no correlation between the amount of tube formation and tumoroid-secreted VEGF-A. Our data suggests that the CACC assay recapitulates the complexity of tumour angiogenesis, and when clinically verified, could prove a valuable tool to quantify sensitivity towards different anti-angiogenic agents.
Collapse
Affiliation(s)
| | - Nabi Mousavi
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Haoche Wei
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Lucy Harvey
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | | | | | | - Klaus Qvortrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Henrik Harling
- 2cureX, Symbion, Copenhagen, Denmark
- Department of Digestive Diseases, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Harry Mellor
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | |
Collapse
|
6
|
Association between [ 68Ga]NODAGA-RGDyK uptake and dynamics of angiogenesis in a human cell-based 3D model. Mol Biol Rep 2021; 48:5347-5353. [PMID: 34213709 PMCID: PMC8318966 DOI: 10.1007/s11033-021-06513-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 06/25/2021] [Indexed: 12/22/2022]
Abstract
Radiolabeled RGD peptides targeting expression of αvβ3 integrin have been applied to in vivo imaging of angiogenesis. However, there is a need for more information on the quantitative relationships between RGD peptide uptake and the dynamics of angiogenesis. In this study, we sought to measure the binding of [68Ga]NODAGA-RGDyK to αvβ3 integrin in a human cell-based three-dimensional (3D) in vitro model of angiogenesis, and to compare the level of binding with the amount of angiogenesis. Experiments were conducted using a human cell-based 3D model of angiogenesis consisting of co-culture of human adipose stem cells (hASCs) and of human umbilical vein endothelial cells (HUVECs). Angiogenesis was induced with four concentrations (25%, 50%, 75%, and 100%) of growth factor cocktail resulting in a gradual increase in the density of the tubule network. Cultures were incubated with [68Ga]NODAGA-RGDyK for 90 min at 37 °C, and binding of radioactivity was measured by gamma counting and digital autoradiography. The results revealed that tracer binding increased gradually with neovasculature density. In comparison with vessels induced with a growth factor concentration of 25%, the uptake of [68Ga]NODAGA-RGDyK was higher at concentrations of 75% and 100%, and correlated with the amount of neovasculature, as determined by visual evaluation of histological staining. Uptake of [68Ga]NODAGA-RGDyK closely reflected the amount of angiogenesis in an in vitro 3D model of angiogenesis. These results support further evaluation of RGD-based approaches for targeted imaging of angiogenesis.
Collapse
|
7
|
Berry SB, Gower MS, Su X, Seshadri C, Theberge AB. A Modular Microscale Granuloma Model for Immune-Microenvironment Signaling Studies in vitro. Front Bioeng Biotechnol 2020; 8:931. [PMID: 32974300 PMCID: PMC7461927 DOI: 10.3389/fbioe.2020.00931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is one of the most potent infectious diseases in the world, causing more deaths than any other single infectious agent. TB infection is caused by inhalation of Mycobacterium tuberculosis (Mtb) and subsequent phagocytosis and migration into the lung tissue by innate immune cells (e.g., alveolar macrophages, neutrophils, and dendritic cells), resulting in the formation of a fused mass of immune cells known as the granuloma. Considered the pathological hallmark of TB, the granuloma is a complex microenvironment that is crucial for pathogen containment as well as pathogen survival. Disruption of the delicate granuloma microenvironment via numerous stimuli, such as variations in cytokine secretions, nutrient availability, and the makeup of immune cell population, can lead to an active infection. Herein, we present a novel in vitro model to examine the soluble factor signaling between a mycobacterial infection and its surrounding environment. Adapting a newly developed suspended microfluidic platform, known as Stacks, we established a modular microscale infection model containing human immune cells and a model mycobacterial strain that can easily integrate with different microenvironmental cues through simple spatial and temporal "stacking" of each module of the platform. We validate the establishment of suspended microscale (4 μL) infection cultures that secrete increased levels of proinflammatory factors IL-6, VEGF, and TNFα upon infection and form 3D aggregates (granuloma model) encapsulating the mycobacteria. As a proof of concept to demonstrate the capability of our platform to examine soluble factor signaling, we cocultured an in vitro angiogenesis model with the granuloma model and quantified morphology changes in endothelial structures as a result of culture conditions (P < 0.05 when comparing infected vs. uninfected coculture systems). We envision our modular in vitro granuloma model can be further expanded and adapted for studies focusing on the complex interplay between granulomatous structures and their surrounding microenvironment, as well as a complementary tool to augment in vivo signaling and mechanistic studies.
Collapse
Affiliation(s)
- Samuel B. Berry
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Maia S. Gower
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Xiaojing Su
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Chetan Seshadri
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, WA, United States
- Department of Urology, University of Washington, Seattle, WA, United States
| |
Collapse
|
8
|
Davidov T, Efraim Y, Dahan N, Baruch L, Machluf M. Porcine arterial ECM hydrogel: Designing an in vitro angiogenesis model for long-term high-throughput research. FASEB J 2020; 34:7745-7758. [PMID: 32337805 DOI: 10.1096/fj.202000264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
The field of angiogenesis research provides deep understanding regarding this important process, which plays fundamental roles in tissue development and different abnormalities. In vitro models offer the advantages of low-cost high-throughput research of angiogenesis while sparing animal lives, and enabling the use of human cells. Nevertheless, prevailing in vitro models lack stability and are limited to a few days' assays. This study, therefore, examines the hypothesis that closely mimicking the vascular microenvironment can more reliably support longer angiogenesis processes in vitro. To this end, porcine arterial extracellular matrix (paECM)- a key component of blood vessels-was isolated and processed into a thermally induced hydrogel and characterized in terms of composition, structure, and mechanical properties, thus confirming the preservation of important characteristics of arterial extracellular matrix. This unique hydrogel was further tailored into a three-dimensional model of angiogenesis using endothelial cells and supporting cells, in a configuration that allows high-throughput quantitative analysis of cell viability and proliferation, cell migration, and apoptosis, thus revealing the advantages of paECM over frequently used biomaterials. Markedly, when applied with well-known effectors of angiogenesis, the model measures reflected the expected response, hence validating its efficacy and establishing its potential as a promising tool for the research of angiogenesis.
Collapse
Affiliation(s)
- Tzila Davidov
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yael Efraim
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nitsan Dahan
- Infrastructure Unit, Life Science and Engineering Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Limor Baruch
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Marcelle Machluf
- Faculty of Biotechnology & Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
9
|
Ratushnyy A, Ezdakova M, Buravkova L. Secretome of Senescent Adipose-Derived Mesenchymal Stem Cells Negatively Regulates Angiogenesis. Int J Mol Sci 2020; 21:ijms21051802. [PMID: 32151085 PMCID: PMC7084202 DOI: 10.3390/ijms21051802] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Nowadays, paracrine regulation is considered as a major tool of mesenchymal stem cell (MSC) involvement in tissue repair and renewal in adults. Aging results in alteration of tissue homeostasis including neovascularization. In this study, we examined the influence of replicative senescence on the angiogenic potential of adipose-derived MSCs (ASCs). Angiogenic activity of conditioned medium (CM) from senescent and “young” ASCs was evaluated in chorioallantoic membrane (CAM) assay in ovo using Japanese quail embryos. Also, the formation of capillary-like tubes by human umbilical vein endothelial cells (HUVECs) in 3D basement membrane matrix “Matrigel” and HUVEC migration capacity were analyzed. Multiplex, dot-blot and gene expression analysis were performed to characterize transcription and production of about 100 angiogenesis-associated proteins. The results point to decreased angiogenic potential of senescent ASC secretome in ovo. A number of angiogenesis-associated proteins demonstrated elevation in CM after long-term cultivation. Meanwhile, VEGF (key positive regulator of angiogenesis) did not change transcription level and concentration in CM. Increasing both pro- (FGF-2, uPA, IL-6, IL-8 etc.) and antiangiogenic (IL-4, IP-10, PF4, Activin A, DPPIV etc.) factors was observed. Some proangiogenic genes were downregulated (IGF1, MMP1, TGFB3, PDGFRB, PGF). Senescence-associated secretory phenotype (SASP) modifications after long-term cultivation lead to attenuation of angiogenic potential of ASC.
Collapse
|
10
|
Reconfigurable open microfluidics for studying the spatiotemporal dynamics of paracrine signalling. Nat Biomed Eng 2019; 3:830-841. [PMID: 31427781 DOI: 10.1038/s41551-019-0421-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/04/2019] [Indexed: 12/17/2022]
Abstract
The study of intercellular signalling networks requires organotypic microscale systems that facilitate the culture, conditioning and manipulation of cells. Here, we describe a reconfigurable microfluidic cell-culture system that facilitates the assembly of three-dimensional tissue models by stacking layers that contain preconditioned microenvironments. By using principles of open and suspended microfluidics, the Stacks system is easily assembled or disassembled to provide spatial and temporal manoeuvrability in two-dimensional and three-dimensional assays of multiple cell types, enabling the modelling of sequential paracrine-signalling events, such as tumour-cell-mediated differentiation of macrophages and macrophage-facilitated angiogenesis. We used Stacks to recapitulate the in vivo observation that different prostate cancer tissues polarize macrophages with distinct gene-expression profiles of pro-inflammatory and anti-inflammatory cytokines. Stacks also enabled us to show that these two types of macrophages signal distinctly to endothelial cells, leading to blood vessels with different morphologies. Our proof-of-concept experiments exemplify how Stacks can efficiently model multicellular interactions and highlight the importance of spatiotemporal specificity in intercellular signalling.
Collapse
|
11
|
Abstract
The more than 80,000 chemicals in commerce present a challenge for hazard assessments that toxicity testing in the 21st century strives to address through high-throughput screening (HTS) assays. Assessing chemical effects on human development adds an additional layer of complexity to the screening, with a need to capture complex and dynamic events essential for proper embryo-fetal development. HTS data from ToxCast/Tox21 informs systems toxicology models, which incorporate molecular targets and biological pathways into mechanistic models describing the effects of chemicals on human cells, 3D organotypic culture models, and small model organisms. Adverse Outcome Pathways (AOPs) provide a useful framework for integrating the evidence derived from these in silico and in vitro systems to inform chemical hazard characterization. To illustrate this formulation, we have built an AOP for developmental toxicity through a mode of action linked to embryonic vascular disruption (Aop43). Here, we review the model for quantitative prediction of developmental vascular toxicity from ToxCast HTS data and compare the HTS results to functional vascular development assays in complex cell systems, virtual tissues, and small model organisms. ToxCast HTS predictions from several published and unpublished assays covering different aspects of the angiogenic cycle were generated for a test set of 38 chemicals representing a range of putative vascular disrupting compounds (pVDCs). Results boost confidence in the capacity to predict adverse developmental outcomes from HTS in vitro data and model computational dynamics for in silico reconstruction of developmental systems biology. Finally, we demonstrate the integration of the AOP and developmental systems toxicology to investigate the unique modes of action of two angiogenesis inhibitors.
Collapse
|
12
|
Nguyen EH, Murphy WL. Customizable biomaterials as tools for advanced anti-angiogenic drug discovery. Biomaterials 2018; 181:53-66. [PMID: 30077137 DOI: 10.1016/j.biomaterials.2018.07.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The inhibition of angiogenesis is a critical element of cancer therapy, as cancer vasculature contributes to tumor expansion. While numerous drugs have proven to be effective at disrupting cancer vasculature, patient survival has not significantly improved as a result of anti-angiogenic drug treatment. Emerging evidence suggests that this is due to a combination of unintended side effects resulting from the application of anti-angiogenic compounds, including angiogenic rebound after treatment and the activation of metastasis in the tumor. There is currently a need to better understand the far-reaching effects of anti-angiogenic drug treatments in the context of cancer. Numerous innovations and discoveries in biomaterials design and tissue engineering techniques are providing investigators with tools to develop physiologically relevant vascular models and gain insights into the holistic impact of drug treatments on tumors. This review examines recent advances in the design of pro-angiogenic biomaterials, specifically in controlling integrin-mediated cell adhesion, growth factor signaling, mechanical properties and oxygen tension, as well as the implementation of pro-angiogenic materials into sophisticated co-culture models of cancer vasculature.
Collapse
Affiliation(s)
- Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
13
|
Angiogenic capacity in pre-eclampsia and uncomplicated pregnancy estimated by assay of angiogenic proteins and an in vitro vasculogenesis/angiogenesis test. Angiogenesis 2018; 22:67-74. [PMID: 30003436 DOI: 10.1007/s10456-018-9637-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/05/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The purpose of the study was to determine the angiogenic capacity of sera in early and late pregnancy and in umbilical blood serum after childbirth, and to define how angiogenic properties assessed in a functional in vitro test are related to individual angiogenic proteins in six women with pre-eclampsia and in six healthy pregnant controls. METHODS Maternal first and third trimester serum samples, and umbilical blood samples after childbirth, were tested in an in vitro human adipose stromal cell-human umbilical vein endothelial cell (hASC-HUVEC) vasculogenesis/angiogenesis assay. The angiogenic properties of the samples were measured by quantifying tubule formation. Concentrations of total placental growth factor (PlGF), total vascular endothelial growth factor (VEGF), soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sEng) were determined by immunoassay. RESULTS First-trimester maternal sera of both groups had a stimulatory effect on angiogenesis in vitro and levels of angiogenic proteins did not differ between the groups. Third-trimester maternal sera in the pre-eclampsia group had an inhibitory effect on tubule formation, while those from normal pregnancies remained stimulatory. Compared with the first trimester there was a significant change in the concentrations of angiogenic proteins toward an anti-angiogenic state in pre-eclampsia. Umbilical blood serum exhibited strong anti-angiogenic effects without a significant difference between groups. CONCLUSIONS Third-trimester serum of pre-eclamptic patients is anti-angiogenic. This phenomenon is not yet present in the first trimester. Umbilical blood serum shows inhibitory effects on angiogenesis after normal as well as pre-eclamptic pregnancy.
Collapse
|
14
|
Iwata Y, Klaren WD, Lebakken CS, Grimm FA, Rusyn I. High-Content Assay Multiplexing for Vascular Toxicity Screening in Induced Pluripotent Stem Cell-Derived Endothelial Cells and Human Umbilical Vein Endothelial Cells. Assay Drug Dev Technol 2017; 15:267-279. [PMID: 28771372 DOI: 10.1089/adt.2017.786] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells (ECs) play a major role in blood vessel formation and function. While there is longstanding evidence for the potential of chemical exposures to adversely affect EC function and vascular development, the hazard potential of chemicals with respect to vascular effects is not routinely evaluated in safety assessments. Induced pluripotent stem cell (iPSC)-derived ECs promise to provide a physiologically relevant, organotypic culture model that is amenable for high-throughput (HT) EC toxicant screening and may represent a viable alternative to traditional in vitro models, including human umbilical vein endothelial cells (HUVECs). To evaluate the utility of iPSC-ECs for multidimensional HT toxicity profiling of chemicals, both iPSC-ECs and HUVECs were exposed to selected positive (angiogenesis inhibitors, cytotoxic agents) and negative compounds in concentration response for either 16 or 24 h in a 384-well plate format. Furthermore, chemical effects on vascularization were quantified using EC angiogenesis on biological (Geltrex™) and synthetic (SP-105 angiogenesis hydrogel) extracellular matrices. Cellular toxicity was assessed using high-content live cell imaging and the CellTiter-Glo® assay. Assay performance indicated good to excellent assay sensitivity and reproducibility for both cell types investigated. Both iPSC-derived ECs and HUVECs formed tube-like structures on Geltrex™ and hydrogel, an effect that was inhibited by angiogenesis inhibitors and cytotoxic agents in a concentration-dependent manner. The quality of HT assays in HUVECs was generally higher than that in iPSC-ECs. Altogether, this study demonstrates the capability of ECs for comprehensive assessment of the biological effects of chemicals on vasculature in a HT compatible format.
Collapse
Affiliation(s)
- Yasuhiro Iwata
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - William D Klaren
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | | | - Fabian A Grimm
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Ivan Rusyn
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| |
Collapse
|
15
|
Maturation of human pluripotent stem cell derived cardiomyocytes is improved in cardiovascular construct. Cytotechnology 2017; 69:785-800. [PMID: 28397099 PMCID: PMC5595750 DOI: 10.1007/s10616-017-0088-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/17/2017] [Indexed: 11/21/2022] Open
Abstract
In order to translate preclinical data into the clinical studies, relevant in vitro models with structure and key functional properties similar to native human tissue should be used. In vitro cardiac models with vascular structures mimic the highly vascularized myocardium and provide interactions between endothelial cells, stromal cells and cardiomyocytes. Currently, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been shown to present immature morphology and fetal-like electrophysiological properties that may limit their use as physiological test platform. The aim of this study was to develop multicellular in vitro cardiovascular construct modeling human heart tissue. In the cardiovascular construct, hPSC-CMs were cultured with a vascular-like network formed by human foreskin fibroblasts and human umbilical vein endothelial cells that served as a platform in the construct. Cardiomyocyte orientation, maturation, electrophysiological properties and drug responses of the cardiovascular construct were characterized and compared to CM monoculture. hPSC-CMs in cardiovascular construct showed elongated morphology and aligned with the vascular-like network. Electrophysiological properties and calcium metabolism of hPSC-CMs as well as response to E-4031 and adrenaline demonstrated normal physiological behavior. Increased expression of cardiac structural proteins and ion channels in cardiovascular construct compared to CM monoculture were detected. In conclusion, vascular-like network supports the structural and functional maturation of hPSC-CMs. Our results suggest that cardiovascular construct presents more mature in vitro cardiac model compared to CM monoculture and could therefore serve as an advanced test system for cardiac safety and efficacy assessment as well as a model system for biomedical research.
Collapse
|
16
|
The effects of culture conditions on the functionality of efficiently obtained mesenchymal stromal cells from human cord blood. Cytotherapy 2016; 18:423-37. [PMID: 26857232 DOI: 10.1016/j.jcyt.2015.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/02/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Cord blood (CB) is an attractive source of mesenchymal stromal cells (MSCs) because of its abundant availability and ease of collection. However, the success rate of generating CB-MSCs is low. In this study, our aim was to demonstrate the efficiency of our previously described method to obtain MSCs from CB and further characterize them and to study the effects of different culture conditions on MSCs. METHODS CB-MSC cultures were established in low oxygen (3%) conditions on fibronectin in 10% fetal bovine serum containing culture medium supplemented with combinations of growth factors. Cells were characterized for their adipogenic, osteogenic and chondrogenic differentiation capacity; phenotype; and HOX gene expression profile. The functionality of the cells cultured in different media was tested in vitro with angiogenesis and T-cell proliferation assays. RESULTS We demonstrate 87% efficacy in generating MSCs from CB. The established cells had typical MSC characteristics with reduced adipogenic differentiation potential and a unique HOX gene fingerprint. Growth factor-rich medium and a 3% oxygen condition enhanced cell proliferation; however, the growth factor-rich medium had a negative effect on the expression of CD90. Dexamethasone-containing medium improved the capacity of the cells to suppress T-cell proliferation, whereas the cells grown without dexamethasone were more able to support angiogenesis. CONCLUSIONS Our results demonstrate that the composition of expansion medium is critical for the functionality of MSCs and should always be appropriately defined for each purpose.
Collapse
|
17
|
Zanotelli MR, Ardalani H, Zhang J, Hou Z, Nguyen EH, Swanson S, Nguyen BK, Bolin J, Elwell A, Bischel LL, Xie AW, Stewart R, Beebe DJ, Thomson JA, Schwartz MP, Murphy WL. Stable engineered vascular networks from human induced pluripotent stem cell-derived endothelial cells cultured in synthetic hydrogels. Acta Biomater 2016; 35:32-41. [PMID: 26945632 PMCID: PMC4829480 DOI: 10.1016/j.actbio.2016.03.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Here, we describe an in vitro strategy to model vascular morphogenesis where human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) are encapsulated in peptide-functionalized poly(ethylene glycol) (PEG) hydrogels, either on standard well plates or within a passive pumping polydimethylsiloxane (PDMS) tri-channel microfluidic device. PEG hydrogels permissive towards cellular remodeling were fabricated using thiol-ene photopolymerization to incorporate matrix metalloproteinase (MMP)-degradable crosslinks and CRGDS cell adhesion peptide. Time lapse microscopy, immunofluorescence imaging, and RNA sequencing (RNA-Seq) demonstrated that iPSC-ECs formed vascular networks through mechanisms that were consistent with in vivo vasculogenesis and angiogenesis when cultured in PEG hydrogels. Migrating iPSC-ECs condensed into clusters, elongated into tubules, and formed polygonal networks through sprouting. Genes upregulated for iPSC-ECs cultured in PEG hydrogels relative to control cells on tissue culture polystyrene (TCP) surfaces included adhesion, matrix remodeling, and Notch signaling pathway genes relevant to in vivo vascular development. Vascular networks with lumens were stable for at least 14days when iPSC-ECs were encapsulated in PEG hydrogels that were polymerized within the central channel of the microfluidic device. Therefore, iPSC-ECs cultured in peptide-functionalized PEG hydrogels offer a defined platform for investigating vascular morphogenesis in vitro using both standard and microfluidic formats. STATEMENT OF SIGNIFICANCE Human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) cultured in synthetic hydrogels self-assemble into capillary networks through mechanisms consistent with in vivo vascular morphogenesis.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | - Hamisha Ardalani
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | - Jue Zhang
- Morgridge Institute for Research, Madison, WI, USA
| | | | - Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | | | | | | | | | - Lauren L Bischel
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | - Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI, USA; Department of Molecular, Cellular, and Developmental Biology, University of California-Santa Barbara, CA, USA
| | - Michael P Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
18
|
Abstract
At present, animal-based models are the major test systems for assessing the tolerability and safety of chemical substances for regulatory purposes, and also for pivotal efficacy testing in pharmaceutical development. In spite of the high genetic similarity between many laboratory animals and humans, animal models are very poor predictors of human health effects and pathophysiological processes. Thus, models and testing strategies that are more relevant to human biology, are needed for these purposes. The best predictability is achieved with human organotypic models that mimic the microenvironment of human tissues. During their development, such models have to be characterised at the structural, genetic and functional levels, and compared to the respective human tissues. Their predictivity should be confirmed by using known reference chemicals with corresponding human data. The use of these methods in safety assessment and biomedical research, combined with the knowledge gained of the underlying biological processes on gene and protein expression, as well as on cellular signalling, will ultimately lead to better human science and animal welfare.
Collapse
Affiliation(s)
- Tuula Heinonen
- Finnish Centre for Alternative Methods, School of Medicine, University of Tampere, Finland
| |
Collapse
|
19
|
Theberge AB, Yu J, Young EWK, Ricke WA, Bushman W, Beebe DJ. Microfluidic multiculture assay to analyze biomolecular signaling in angiogenesis. Anal Chem 2015; 87:3239-46. [PMID: 25719435 PMCID: PMC4405103 DOI: 10.1021/ac503700f] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Angiogenesis (the formation of blood vessels from existing blood vessels) plays a critical role in many diseases such as cancer, benign tumors, and macular degeneration. There is a need for cell culture methods capable of dissecting the intricate regulation of angiogenesis within the microenvironment of the vasculature. We have developed a microscale cell-based assay that responds to complex pro- and antiangiogenic soluble factors with an in vitro readout for vessel formation. The power of this system over traditional techniques is that we can incorporate the whole milieu of soluble factors produced by cells in situ into one biological readout (vessel formation), even if the identity of the factors is unknown. We have currently incorporated macrophages, endothelial cells, and fibroblasts into the assay, with the potential to include additional cell types in the future. Importantly, the microfluidic platform is simple to operate and multiplex to test drugs targeting angiogenesis in a more physiologically relevant context. As a proof of concept, we tested the effect of an enzyme inhibitor (targeting matrix metalloproteinase 12) on vessel formation; the triculture microfluidic assay enabled us to capture a dose-dependent effect entirely missed in a simplified coculture assay (p < 0.0001). This result underscores the importance of cell-based assays that capture chemical cross-talk occurring between cell types. The microscale dimensions significantly reduce cell consumption compared to conventional well plate platforms, enabling the use of limited primary cells from patients in future investigations and offering the potential to screen therapeutic approaches for individual patients in vitro.
Collapse
Affiliation(s)
- Ashleigh B. Theberge
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jiaquan Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Edmond W. K. Young
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Mechanical & Industrial Engineering, Institute of Biomaterials and Biomedical Engineering, Toronto, Canada, M5S 3G8
| | - William A. Ricke
- Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Wade Bushman
- Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
20
|
Vuorenpää H, Ikonen L, Kujala K, Huttala O, Sarkanen JR, Ylikomi T, Aalto-Setälä K, Heinonen T. Novel in vitro cardiovascular constructs composed of vascular-like networks and cardiomyocytes. In Vitro Cell Dev Biol Anim 2013; 50:275-86. [PMID: 24163159 DOI: 10.1007/s11626-013-9703-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 10/03/2013] [Indexed: 01/17/2023]
Abstract
The interaction between different cardiac cells has shown to be important for critical biological properties including cell survival, proliferation, differentiation and function. The improvement of culture conditions with different cell types and to study their effects on cardiomyocyte viability and functionality is essential. For practical applications including general toxicity testing, drug development and tissue engineering it is important to study whether co-cultures have additional advantages over cardiomyocyte monoculture. Two multicellular in vitro cardiovascular constructs devoid of added biomaterial were developed in this study. In the first construct, neonatal rat cardiomyocytes (CM) were seeded on vascular-like network formed by human umbilical vein endothelial cells (HUVEC) and human adipose stromal cells (hASC). In the second construct, CMs were seeded on vascular-like network formed by HUVECs and human foreskin fibroblasts. The ability of these two vascular-like networks to support the viability and functionality of CMs was analyzed. Different culture media compositions were evaluated to support the development of optimal cardiovascular construct. Our results demonstrate that both vascular-like networks markedly improved CM viability and functionality. In the constructs, co-localization of CMs and vascular-like networks was seen. Multicellular constructs also allowed synchronized contractility of CMs. Serum-free medium supplemented with vascular endothelial growth factor and basic fibroblast growth factor was found to provide the most optimal conditions for cardiovascular construct as an entity. In conclusion, when combining a vascular-like network with CMs, the viability and functionality of CMs was markedly improved. The results suggest that the cardiovascular constructs developed provide a promising new tool for the assessment of toxicological and safety pharmacological effects of compounds in vitro.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- FICAM, Finnish Centre for Alternative Methods, School of Medicine, University of Tampere, Medisiinarinkatu 3, 33014, Tampere, Finland,
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Friis T, Engel AM, Bendiksen CD, Larsen LS, Houen G. Influence of levamisole and other angiogenesis inhibitors on angiogenesis and endothelial cell morphology in vitro. Cancers (Basel) 2013; 5:762-85. [PMID: 24202320 PMCID: PMC3795364 DOI: 10.3390/cancers5030762] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/11/2013] [Accepted: 06/14/2013] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels from existing vessels is required for many physiological processes and for growth of solid tumors. Initiated by hypoxia, angiogenesis involves binding of angiogenic factors to endothelial cell (EC) receptors and activation of cellular signaling, differentiation, migration, proliferation, interconnection and canalization of ECs, remodeling of the extracellular matrix and stabilization of newly formed vessels. Experimentally, these processes can be studied by several in vitro and in vivo assays focusing on different steps in the process. In vitro, ECs form networks of capillary-like tubes when propagated for three days in coculture with fibroblasts. The tube formation is dependent on vascular endothelial growth factor (VEGF) and omission of VEGF from the culture medium results in the formation of clusters of undifferentiated ECs. Addition of angiogenesis inhibitors to the coculture system disrupts endothelial network formation and influences EC morphology in two distinct ways. Treatment with antibodies to VEGF, soluble VEGF receptor, the VEGF receptor tyrosine kinase inhibitor SU5614, protein tyrosine phosphatase inhibitor (PTPI) IV or levamisole results in the formation of EC clusters of variable size. This cluster morphology is a result of inhibited EC differentiation and levamisole can be inferred to influence and block VEGF signaling. Treatment with platelet factor 4, thrombospondin, rapamycin, suramin, TNP-470, salubrinal, PTPI I, PTPI II, clodronate, NSC87877 or non-steriodal anti-inflammatory drugs (NSAIDs) results in the formation of short cords of ECs, which suggests that these inhibitors have an influence on later steps in the angiogenic process, such as EC proliferation and migration. A humanized antibody to VEGF is one of a few angiogenesis inhibitors used clinically for treatment of cancer. Levamisole is approved for clinical treatment of cancer and is interesting with respect to anti-angiogenic activity in vivo since it inhibits ECs in vitro with a morphology resembling that obtained with antibodies to VEGF.
Collapse
Affiliation(s)
- Tina Friis
- Department of Clinical Biochemistry, Immunology and Genetics, Statens Serum Institut, Artillerivej 5, DK-2300 Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
22
|
Kerkelä E, Hakkarainen T, Mäkelä T, Raki M, Kambur O, Kilpinen L, Nikkilä J, Lehtonen S, Ritamo I, Pernu R, Pietilä M, Takalo R, Juvonen T, Bergström K, Kalso E, Valmu L, Laitinen S, Lehenkari P, Nystedt J. Transient proteolytic modification of mesenchymal stromal cells increases lung clearance rate and targeting to injured tissue. Stem Cells Transl Med 2013; 2:510-20. [PMID: 23734061 DOI: 10.5966/sctm.2012-0187] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Systemic infusion of therapeutic cells would be the most practical and least invasive method of administration in many cellular therapies. One of the main obstacles especially in intravenous delivery of cells is a massive cell retention in the lungs, which impairs homing to the target tissue and may decrease the therapeutic outcome. In this study we showed that an alternative cell detachment of mesenchymal stromal/stem cells (MSCs) with pronase instead of trypsin significantly accelerated the lung clearance of the cells and, importantly, increased their targeting to an area of injury. Cell detachment with pronase transiently altered the MSC surface protein profile without compromising cell viability, multipotent cell characteristics, or immunomodulative and angiogenic potential. The transient modification of the cell surface protein profile was sufficient to produce effective changes in cell rolling behavior in vitro and, importantly, in the in vivo biodistribution of the cells in mouse, rat, and porcine models. In conclusion, pronase detachment could be used as a method to improve the MSC lung clearance and targeting in vivo. This may have a major impact on the bioavailability of MSCs in future therapeutic regimes.
Collapse
Affiliation(s)
- Erja Kerkelä
- Advanced Therapies and Product Development, Finnish.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Knudsen TB, Kleinstreuer NC. Disruption of embryonic vascular development in predictive toxicology. ACTA ACUST UNITED AC 2012; 93:312-23. [DOI: 10.1002/bdrc.20223] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Sarkanen JR, Vuorenpää H, Huttala O, Mannerström B, Kuokkanen H, Miettinen S, Heinonen T, Ylikomi T. Adipose Stromal Cell Tubule Network Model Provides a Versatile Tool for Vascular Research and Tissue Engineering. Cells Tissues Organs 2012; 196:385-97. [DOI: 10.1159/000336679] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2012] [Indexed: 11/19/2022] Open
|
25
|
Sarkanen JR, Kaila V, Mannerström B, Räty S, Kuokkanen H, Miettinen S, Ylikomi T. Human Adipose Tissue Extract Induces Angiogenesis and AdipogenesisIn Vitro. Tissue Eng Part A 2012; 18:17-25. [DOI: 10.1089/ten.tea.2010.0712] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Jertta-Riina Sarkanen
- Department of Cell Biology, Medical School, University of Tampere, Tampere, Finland
- FICAM, Finnish Center for Alternative Methods, Medical School, University of Tampere, Tampere, Finland
- Science Center, Tampere University Hospital, Tampere, Finland
| | - Ville Kaila
- Department of Cell Biology, Medical School, University of Tampere, Tampere, Finland
| | - Bettina Mannerström
- Science Center, Tampere University Hospital, Tampere, Finland
- Adult Stem Cells, Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, Tampere, Finland
| | - Sari Räty
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Hannu Kuokkanen
- Department of Plastic Surgery, Tampere University Hospital, Tampere, Finland
| | - Susanna Miettinen
- Science Center, Tampere University Hospital, Tampere, Finland
- Adult Stem Cells, Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, Tampere, Finland
| | - Timo Ylikomi
- Department of Cell Biology, Medical School, University of Tampere, Tampere, Finland
- FICAM, Finnish Center for Alternative Methods, Medical School, University of Tampere, Tampere, Finland
- Department of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
26
|
Kleinstreuer NC, Judson RS, Reif DM, Sipes NS, Singh AV, Chandler KJ, Dewoskin R, Dix DJ, Kavlock RJ, Knudsen TB. Environmental impact on vascular development predicted by high-throughput screening. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:1596-603. [PMID: 21788198 PMCID: PMC3226499 DOI: 10.1289/ehp.1103412] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 07/25/2011] [Indexed: 05/07/2023]
Abstract
BACKGROUND Understanding health risks to embryonic development from exposure to environmental chemicals is a significant challenge given the diverse chemical landscape and paucity of data for most of these compounds. High-throughput screening (HTS) in the U.S. Environmental Protection Agency (EPA) ToxCast™ project provides vast data on an expanding chemical library currently consisting of > 1,000 unique compounds across > 500 in vitro assays in phase I (complete) and Phase II (under way). This public data set can be used to evaluate concentration-dependent effects on many diverse biological targets and build predictive models of prototypical toxicity pathways that can aid decision making for assessments of human developmental health and disease. OBJECTIVE We mined the ToxCast phase I data set to identify signatures for potential chemical disruption of blood vessel formation and remodeling. METHODS ToxCast phase I screened 309 chemicals using 467 HTS assays across nine assay technology platforms. The assays measured direct interactions between chemicals and molecular targets (receptors, enzymes), as well as downstream effects on reporter gene activity or cellular consequences. We ranked the chemicals according to individual vascular bioactivity score and visualized the ranking using ToxPi (Toxicological Priority Index) profiles. RESULTS Targets in inflammatory chemokine signaling, the vascular endothelial growth factor pathway, and the plasminogen-activating system were strongly perturbed by some chemicals, and we found positive correlations with developmental effects from the U.S. EPA ToxRefDB (Toxicological Reference Database) in vivo database containing prenatal rat and rabbit guideline studies. We observed distinctly different correlative patterns for chemicals with effects in rabbits versus rats, despite derivation of in vitro signatures based on human cells and cell-free biochemical targets, implying conservation but potentially differential contributions of developmental pathways among species. Follow-up analysis with antiangiogenic thalidomide analogs and additional in vitro vascular targets showed in vitro activity consistent with the most active environmental chemicals tested here. CONCLUSIONS We predicted that blood vessel development is a target for environmental chemicals acting as putative vascular disruptor compounds (pVDCs) and identified potential species differences in sensitive vascular developmental pathways.
Collapse
Affiliation(s)
- Nicole C Kleinstreuer
- National Center for Computational Toxiciology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|