1
|
Bertucci T, Kakarla S, Winkelman MA, Lane K, Stevens K, Lotz S, Grath A, James D, Temple S, Dai G. Direct differentiation of human pluripotent stem cells into vascular network along with supporting mural cells. APL Bioeng 2023; 7:036107. [PMID: 37564277 PMCID: PMC10411996 DOI: 10.1063/5.0155207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
During embryonic development, endothelial cells (ECs) undergo vasculogenesis to form a primitive plexus and assemble into networks comprised of mural cell-stabilized vessels with molecularly distinct artery and vein signatures. This organized vasculature is established prior to the initiation of blood flow and depends on a sequence of complex signaling events elucidated primarily in animal models, but less studied and understood in humans. Here, we have developed a simple vascular differentiation protocol for human pluripotent stem cells that generates ECs, pericytes, and smooth muscle cells simultaneously. When this protocol is applied in a 3D hydrogel, we demonstrate that it recapitulates the dynamic processes of early human vessel formation, including acquisition of distinct arterial and venous fates, resulting in a vasculogenesis angiogenesis model plexus (VAMP). The VAMP captures the major stages of vasculogenesis, angiogenesis, and vascular network formation and is a simple, rapid, scalable model system for studying early human vascular development in vitro.
Collapse
Affiliation(s)
| | - Shravani Kakarla
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Max A. Winkelman
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Keith Lane
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | | | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | - Alexander Grath
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Daylon James
- Weill Cornell Medicine, New York, New York 10065, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | - Guohao Dai
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| |
Collapse
|
2
|
Fitzgerald KN, Motzer RJ, Lee CH. Adjuvant therapy options in renal cell carcinoma - targeting the metastatic cascade. Nat Rev Urol 2023; 20:179-193. [PMID: 36369389 PMCID: PMC10921989 DOI: 10.1038/s41585-022-00666-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2022] [Indexed: 11/13/2022]
Abstract
Localized renal cell carcinoma (RCC) is primarily managed with nephrectomy, which is performed with curative intent. However, disease recurs in ~20% of patients. Treatment with adjuvant therapies is used after surgery with the intention of curing additional patients by disrupting the establishment, maturation or survival of micrometastases, processes collectively referred to as the metastatic cascade. Immune checkpoint inhibitors and vascular endothelial growth factor receptor (VEGFR)-targeting tyrosine kinase inhibitors (TKIs) have shown efficacy in the treatment of metastatic RCC, increasing the interest in the utility of these agents in the adjuvant setting. Pembrolizumab, an inhibitor of the immune checkpoint PD1, is now approved by the FDA and is under review by European regulatory agencies for the adjuvant treatment of patients with localized resected clear cell RCC based on the results of the KEYNOTE-564 trial. However, the optimal use of immunotherapy and VEGFR-targeting TKIs for adjuvant treatment of RCC is not completely understood. These agents disrupt the metastatic cascade at multiple steps, providing biological rationale for further investigating the applications of these therapeutics in the adjuvant setting. Clinical trials to evaluate adjuvant therapeutics in RCC are ongoing, and clinical considerations must guide the practical use of immunotherapy and TKI agents for the adjuvant treatment of localized resected RCC.
Collapse
Affiliation(s)
- Kelly N Fitzgerald
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
3
|
Kaarj K, Madias M, Akarapipad P, Cho S, Yoon JY. Paper-based in vitro tissue chip for delivering programmed mechanical stimuli of local compression and shear flow. J Biol Eng 2020; 14:20. [PMID: 32742306 PMCID: PMC7385864 DOI: 10.1186/s13036-020-00242-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT Mechanical stimuli play important roles on the growth, development, and behavior of tissue. A simple and novel paper-based in vitro tissue chip was developed that can deliver two types of mechanical stimuli-local compression and shear flow-in a programmed manner. Rat vascular endothelial cells (RVECs) were patterned on collagen-coated nitrocellulose paper to create a tissue chip. Localized compression and shear flow were introduced by simply tapping and bending the paper chip in a programmed manner, utilizing an inexpensive servo motor controlled by an Arduino microcontroller and powered by batteries. All electrical compartments and a paper-based tissue chip were enclosed in a single 3D-printed enclosure, allowing the whole device to be independently placed within an incubator. This simple device effectively simulated in vivo conditions and induced successful RVEC migration in as early as 5 h. The developed device provides an inexpensive and flexible alternative for delivering mechanical stimuli to other in vitro tissue models. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Kattika Kaarj
- Department of Biosystems Engineering, The University of Arizona, Tucson, AZ USA
| | - Marianne Madias
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ USA
| | | | - Soohee Cho
- Department of Biosystems Engineering, The University of Arizona, Tucson, AZ USA
| | - Jeong-Yeol Yoon
- Department of Biosystems Engineering, The University of Arizona, Tucson, AZ USA
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ USA
| |
Collapse
|
4
|
Farooq M, Abutaha N, Mahboob S, Baabbad A, Almoutiri ND, Wadaan MAAM. Investigating the antiangiogenic potential of Rumex vesicarius (humeidh), anticancer activity in cancer cell lines and assessment of developmental toxicity in zebrafish embryos. Saudi J Biol Sci 2020; 27:611-622. [PMID: 32210679 PMCID: PMC6997907 DOI: 10.1016/j.sjbs.2019.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022] Open
Abstract
Recent trends in anticancer therapy is to use therapeutic agents which not only kill the cancer cell, but are less toxic to surrounding normal cells/tissue. One approach is to cut the nutrient supply to growing tumor cells, by blocking the formation of new blood vessels around the tumor. As the phytochemicals and botanical crude extracts have proven their efficacy as natural antiangiogenic agents with minimum toxicities, there is need to explore varieties of medicinal plants for novel antiangiogenic compounds. Rumex vesicarius L. (Humeidh), is an annual herbal plant with proven medicinal values. The antiangiogenic potential, and developmental toxicity of humeidh in experimental animal models has never been studied before. The crude extracts were prepared from the roots, stems, leaves and flowers of Rumex vesicarius L. in methanol, chloroform, ethyl acetate and n-hexane. The developmental toxicity screening in zebrafish embryos, has revealed that Rumex vesicarius was not toxic to zebrafish embryos. The chloroform stem extract showed significant level of antiangiogenic activity in zebrafish angiogenic assay on a dose dependent manner. Thirty five (35) bioactive compounds were identified by gas chromatography mass spectrophotometry (GC–MS) analysis in the stem extract of Rumex vesicarius. Propanoic acid, 2-[(trimethylsilyl)oxy]-, trimethylsilyl ester, Butane, 1,2,3-tris(trimethylsiloxy), and Butanedioic acid, bis(trimethylsilyl) ester were identified as major compound present in the stem of R. vasicarius. The anticancer activity of roots, stem, leaves and flowers crude extract was evaluated in human breast cancer (MCF7), human colon carcinoma (Lovo, and Caco-2), human hepatocellular carcinoma (HepG2) cell lines. Most of the crude extracts did not show significant level of cytotoxicity in tested cancer cells line, except, chloroform extract of stem which exhibited strong anticancer activity in all tested cancer cells with IC50 values in micro molar range. Based on these results, it is recommended that formulation prepared from R. vesicarius can further be tested in clinical trials in order to explore its therapeutic potential as an effective and safe natural anticancer product.
Collapse
Affiliation(s)
- Muhammad Farooq
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Nael Abutaha
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Shahid Mahboob
- Department of Zoology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Almohannad Baabbad
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Nawaf D Almoutiri
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | | |
Collapse
|
5
|
Peloquin GL, Johnston L, Damarla M, Damico RL, Hassoun PM, Kolb TM. SU5416 does not attenuate early RV angiogenesis in the murine chronic hypoxia PH model. Respir Res 2019; 20:123. [PMID: 31208454 PMCID: PMC6580559 DOI: 10.1186/s12931-019-1079-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Right ventricular (RV) angiogenesis has been associated with adaptive myocardial remodeling in pulmonary hypertension (PH), though molecular regulators are poorly defined. Endothelial cell VEGFR-2 is considered a "master regulator" of angiogenesis in other models, and the small molecule VEGF receptor tyrosine kinase inhibitor SU5416 is commonly used to generate PH in rodents. We hypothesized that SU5416, through direct effects on cardiac endothelial cell VEGFR-2, would attenuate RV angiogenesis in a murine model of PH. METHODS C57 BL/6 mice were exposed to chronic hypoxia (CH-PH) to generate PH and stimulate RV angiogenesis. SU5416 (20 mg/kg) or vehicle were administered at the start of the CH exposure, and weekly thereafter. Angiogenesis was measured after one week of CH-PH using a combination of unbiased stereological measurements and flow cytometry-based quantification of myocardial endothelial cell proliferation. In complementary experiments, primary cardiac endothelial cells from C57 BL/6 mice were exposed to recombinant VEGF (50 ng/mL) or grown on Matrigel in the presence of SU5416 (5 μM) or vehicle. RESULT SU5416 directly inhibited VEGF-mediated ERK phosphorylation, cell proliferation, and Kdr transcription, but not Matrigel tube formation in primary murine cardiac endothelial cells in vitro. SU5416 did not inhibit CH-PH induced RV angiogenesis, endothelial cell proliferation, or RV hypertrophy in vivo, despite significantly altering the expression profile of genes involved in angiogenesis. CONCLUSIONS These findings demonstrate that SU5416 directly inhibited VEGF-induced proliferation of murine cardiac endothelial cells but does not attenuate CH-PH induced RV angiogenesis or myocardial remodeling in vivo.
Collapse
Affiliation(s)
- Grace L Peloquin
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, NYU Langone Health, New York, NY, USA
| | - Laura Johnston
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, 5th Floor, Baltimore, MD, 21205, USA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, 5th Floor, Baltimore, MD, 21205, USA
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, 5th Floor, Baltimore, MD, 21205, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, 5th Floor, Baltimore, MD, 21205, USA
| | - Todd M Kolb
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, 5th Floor, Baltimore, MD, 21205, USA.
| |
Collapse
|
6
|
Benning L, Gutzweiler L, Tröndle K, Riba J, Zengerle R, Koltay P, Zimmermann S, Stark GB, Finkenzeller G. Assessment of hydrogels for bioprinting of endothelial cells. J Biomed Mater Res A 2017; 106:935-947. [PMID: 29119674 DOI: 10.1002/jbm.a.36291] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 10/13/2017] [Accepted: 11/02/2017] [Indexed: 11/10/2022]
Abstract
In tissue engineering applications, vascularization can be accomplished by coimplantation of tissue forming cells and endothelial cells (ECs), whereby the latter are able to form functional blood vessels. The use of three-dimensional (3D) bioprinting technologies has the potential to improve the classical tissue engineering approach because these will allow the generation of scaffolds with high spatial control of endothelial cell allocation. This study focuses on a side by side comparison of popular commercially available bioprinting hydrogels (Matrigel, fibrin, collagen, gelatin, agarose, Pluronic F-127, alginate, and alginate/gelatin) in the context of their physicochemical parameters, their swelling/degradation characteristics, their biological effects on vasculogenesis-related EC parameters and their printability. The aim of this study was to identify the most suitable hydrogel or hydrogel combination for inkjet printing of ECs to build prevascularized tissue constructs. Most tested hydrogels displayed physicochemical characteristics suitable for inkjet printing. However, Pluronic F-127 and the alginate/gelatin blend were rapidly degraded when incubated in cell culture medium. Agarose, Pluronic F-127, alginate and alginate/gelatin hydrogels turned out to be unsuitable for bioprinting of ECs because of their non-adherent properties and/or their incapability to support EC proliferation. Gelatin was able to support EC proliferation and viability but was unable to support endothelial cell sprouting. Our experiments revealed fibrin and collagen to be most suitable for bioprinting of ECs, because these hydrogels showed acceptable swelling/degradation characteristics, supported vasculogenesis-related EC parameters and showed good printability. Moreover, ECs in constructs of preformed spheroids survived the printing process and formed capillary-like cords. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 935-947, 2018.
Collapse
Affiliation(s)
- Leo Benning
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ludwig Gutzweiler
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany
| | - Kevin Tröndle
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany
| | - Julian Riba
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany.,Hahn-Schickard, Georges-Koehler-Allee 103, Freiburg, 79110, Germany.,FIT - Freiburg Centre for Interactive Materials and Bioinspired Technologies, University of Freiburg, Georges-Koehler-Allee 105, Freiburg, 79110, Germany
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg, 79110, Germany
| | - G Björn Stark
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Spheroids as vascularization units: From angiogenesis research to tissue engineering applications. Biotechnol Adv 2017; 35:782-791. [DOI: 10.1016/j.biotechadv.2017.07.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 02/08/2023]
|
8
|
Tevis KM, Colson YL, Grinstaff MW. Embedded Spheroids as Models of the Cancer Microenvironment. ADVANCED BIOSYSTEMS 2017; 1:1700083. [PMID: 30221187 PMCID: PMC6135264 DOI: 10.1002/adbi.201700083] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To more accurately study the complex mechanisms behind cancer invasion, progression, and response to treatment, researchers require models that replicate both the multicellular nature and 3D stromal environment present in an in vivo tumor. Multicellular aggregates (i.e., spheroids) embedded in an extracellular matrix mimic are a prevalent model. Recently, quantitative metrics that fully utilize the capability of spheroids are described along with conventional experiments, such as invasion into a matrix, to provide additional details and insights into the underlying cancer biology. The review begins with a discussion of the salient features of the tumor microenvironment, introduces the early work on non-embedded spheroids as tumor models, and then concentrates on the successes achieved with the study of embedded spheroids. Examples of studies include cell movement, drug response, tumor cellular heterogeneity, stromal effects, and cancer progression. Additionally, new methodologies and those borrowed from other research fields (e.g., vascularization and tissue engineering) are highlighted that expand the capability of spheroids to aid future users in designing their cancer-related experiments. The convergence of spheroid research among the various fields catalyzes new applications and leads to a natural synergy. Finally, the review concludes with a reflection and future perspectives for cancer spheroid research.
Collapse
Affiliation(s)
- Kristie M. Tevis
- Departments of Biomedical Engineering, Chemistry, and Medicine, Metcalf Center for Science and Engineering, Boston University, Boston, MA 02215
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02215
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Metcalf Center for Science and Engineering, Boston University, Boston, MA 02215
| |
Collapse
|
9
|
Hirabayashi T, Takahashi H, Watanabe M, Tachibana T. Establishment and characterization of a squamous cell carcinoma cell line, designated hZK-1, derived from a metastatic lymph node tumor of the tongue. Hum Cell 2017; 30:319-326. [PMID: 28260147 DOI: 10.1007/s13577-017-0165-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/17/2017] [Indexed: 12/18/2022]
Abstract
The hZK-1 cell line was successfully established from the metastatic foci of a lymph node of an 82-year-old Japanese woman with squamous cell carcinoma of the tongue. The pathological diagnosis of the tumor was moderately to well-differentiated squamous cell carcinoma. The hZK-1 cells were angular in shape, and had neoplastic and pleomorphic features. Adjacent hZK-1 cells were joined by desmosomes and well-developed microvilli, and many free ribosomes were observed in the cytoplasm. The doubling time of the hZK-1 cells was approximately 36, 33, and 29 h at the 10th, 20th, and 30th passages, respectively. The cell line was shown to be triploid, with a chromosomal distribution of 75-80. Immunocytochemical staining of the hZK-1 cells revealed cytokeratin (CK) 17-, Ki67-, and p53-positive staining, and negative staining for CK13. The hZK-1 cells were negative for human papillomavirus (HPV)-16 or-18 infection. Grafting was not successful when the hZK-1 cells were transplanted into the subcutis of SCID mice. The hZK-1 cells (2 × 106 cells/3 ml of growth medium) secreted vascular endothelial growth factor (VEGF) that reached a concentration of 2.6 ng/ml media after 3 days of culture. Hypoxia enhanced cellular HIF-1α expression and VEGF secretion in hZK-1 cells. The HIF-1α inhibitor YC-1 partially inhibited hypoxia-induced VEGF secretion in ZK-1 cells. The reverse transcription-polymerase chain reaction (RT-PCR) results revealed that the expression of CK17, Ki67, and p53 was elevated in the hZK-1 cells. hZK-1 cells were not sensitive to CDDP, TXT, 5-FU, or a mixture of these three anti-tumor agents.
Collapse
Affiliation(s)
- Tetsuya Hirabayashi
- Department of Anatomy, The Jikei University School of Medicine, 3-25-8 Nishishinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Haruka Takahashi
- Department of Oral and Maxillofacial Surgery, Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-1500, Japan.,Department of NDU Life Sciences, School of Life Dentistry, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan
| | - Miho Watanabe
- Department of Oral and Maxillofacial Surgery, Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-1500, Japan.,Department of NDU Life Sciences, School of Life Dentistry, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan
| | - Toshiaki Tachibana
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
10
|
Belair DG, Schwartz MP, Knudsen T, Murphy WL. Human iPSC-derived endothelial cell sprouting assay in synthetic hydrogel arrays. Acta Biomater 2016; 39:12-24. [PMID: 27181878 PMCID: PMC5228278 DOI: 10.1016/j.actbio.2016.05.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023]
Abstract
UNLABELLED Activation of vascular endothelial cells (ECs) by growth factors initiates a cascade of events during angiogenesis in vivo consisting of EC tip cell selection, sprout formation, EC stalk cell proliferation, and ultimately vascular stabilization by support cells. Although EC functional assays can recapitulate one or more aspects of angiogenesis in vitro, they are often limited by undefined substrates and lack of dependence on key angiogenic signaling axes. Here, we designed and characterized a chemically-defined model of endothelial sprouting behavior in vitro using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs). We rapidly encapsulated iPSC-ECs at high density in poly(ethylene glycol) (PEG) hydrogel spheres using thiol-ene chemistry and subsequently encapsulated cell-dense hydrogel spheres in a cell-free hydrogel layer. The hydrogel sprouting array supported pro-angiogenic phenotype of iPSC-ECs and supported growth factor-dependent proliferation and sprouting behavior. iPSC-ECs in the sprouting model responded appropriately to several reference pharmacological angiogenesis inhibitors of vascular endothelial growth factor, NF-κB, matrix metalloproteinase-2/9, protein kinase activity, and β-tubulin, which confirms their functional role in endothelial sprouting. A blinded screen of 38 putative vascular disrupting compounds from the US Environmental Protection Agency's ToxCast library identified six compounds that inhibited iPSC-EC sprouting and five compounds that were overtly cytotoxic to iPSC-ECs at a single concentration. The chemically-defined iPSC-EC sprouting model (iSM) is thus amenable to enhanced-throughput screening of small molecular libraries for effects on angiogenic sprouting and iPSC-EC toxicity assessment. STATEMENT OF SIGNIFICANCE Angiogenesis assays that are commonly used for drug screening and toxicity assessment applications typically utilize natural substrates like Matrigel(TM) that are difficult to spatially pattern, costly, ill-defined, and may exhibit lot-to-lot variability. Herein, we describe a novel angiogenic sprouting assay using chemically-defined, bioinert poly(ethylene glycol) hydrogels functionalized with biomimetic peptides to promote cell attachment and degradation in a reproducible format that may mitigate the need for natural substrates. The quantitative assay of angiogenic sprouting here enables precise control over the initial conditions and can be formulated into arrays for screening. The sprouting assay here was dependent on key angiogenic signaling axes in a screen of angiogenesis inhibitors and a blinded screen of putative vascular disrupting compounds from the US-EPA.
Collapse
Affiliation(s)
- David G Belair
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael P Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas Knudsen
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Material Science Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Human vascular tissue models formed from human induced pluripotent stem cell derived endothelial cells. Stem Cell Rev Rep 2016; 11:511-25. [PMID: 25190668 DOI: 10.1007/s12015-014-9549-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Here we describe a strategy to model blood vessel development using a well-defined induced pluripotent stem cell-derived endothelial cell type (iPSC-EC) cultured within engineered platforms that mimic the 3D microenvironment. The iPSC-ECs used here were first characterized by expression of endothelial markers and functional properties that included VEGF responsiveness, TNF-α-induced upregulation of cell adhesion molecules (MCAM/CD146; ICAM1/CD54), thrombin-dependent barrier function, shear stress-induced alignment, and 2D and 3D capillary-like network formation in Matrigel. The iPSC-ECs also formed 3D vascular networks in a variety of engineering contexts, yielded perfusable, interconnected lumen when co-cultured with primary human fibroblasts, and aligned with flow in microfluidics devices. iPSC-EC function during tubule network formation, barrier formation, and sprouting was consistent with that of primary ECs, and the results suggest a VEGF-independent mechanism for sprouting, which is relevant to therapeutic anti-angiogenesis strategies. Our combined results demonstrate the feasibility of using a well-defined, stable source of iPSC-ECs to model blood vessel formation within a variety of contexts using standard in vitro formats.
Collapse
|
12
|
Benien P, Swami A. 3D tumor models: history, advances and future perspectives. Future Oncol 2014; 10:1311-27. [DOI: 10.2217/fon.13.274] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT: Evaluation of cancer therapeutics by utilizing 3D tumor models, before clinical studies, could be more advantageous than conventional 2D tumor models (monolayer cultures). The 3D systems mimic the tumor microenvironment more closely than 2D systems. The following review discusses the various 3D tumor models present today with the advantages and limitations of each. 3D tumor models replicate the elements of a tumor microenvironment such as hypoxia, necrosis, angiogenesis and cell adhesion. The review introduces application of techniques such as microfluidics, imaging and tissue engineering to improve the 3D tumor models. Despite their tremendous potential to better screen chemotherapeutics, 3D tumor models still have a long way to go before they are used commonly as in vitro tumor models in pharmaceutical industrial research.
Collapse
Affiliation(s)
| | - Archana Swami
- Department of Anesthesiology, Brigham & Women’s Hospital Boston, MA 02115, USA
| |
Collapse
|
13
|
Zhou TT, He L, Yan M, Zhang LY, He JG, Rao XP. Tyrosine kinase inhibitory activity of dehydroabietylamine derivatives tested by homogeneous time-resolved fluorescence based high throughput screening model. Chin J Nat Med 2013; 11:506-13. [PMID: 24359775 DOI: 10.1016/s1875-5364(13)60092-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Indexed: 10/25/2022]
Abstract
Protein tyrosine kinases (PTKs) are attractive targets in searching for therapeutic agents against many diseases. In this study, a series of dehydroabietylamine derivatives were first determined to show PTK inhibitory activity using a high-throughput screening (HTS) method based on homogeneous time-resolved fluorescence (HTRF) technology. The structure-activity relationships of the dehydroabietylamine derivatives were established, and it was found that the compounds with a nitrogen-containing side chain had better inhibitory activity. Further studies showed that the compounds substituted with halogen in the phenyl ring resulted in higher inhibitory activity on the epidermal growth factor receptor (EGFR), and can be a guide to modify the structure of dehydroabietylamine derivatives. Dehydroabietylamine derivatives might be a new class of multi-targeted and effective PTK inhibitors with structure modifications.
Collapse
Affiliation(s)
- Tao-Tao Zhou
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| | - Ming Yan
- National Drug Screening Laboratory, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-Yong Zhang
- National Drug Screening Laboratory, China Pharmaceutical University, Nanjing 210009, China
| | - Jian-Guo He
- Department of Neurosurgery, Chongqing Red Cross Hospital, Chongqing 400020, China
| | - Xiao-Ping Rao
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China
| |
Collapse
|
14
|
Mohan N, Karmakar S, Banik NL, Ray SK. SU5416 and EGCG work synergistically and inhibit angiogenic and survival factors and induce cell cycle arrest to promote apoptosis in human malignant neuroblastoma SH-SY5Y and SK-N-BE2 cells. Neurochem Res 2011; 36:1383-96. [PMID: 21472456 DOI: 10.1007/s11064-011-0463-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2011] [Indexed: 12/20/2022]
Abstract
Malignant neuroblastomas are solid tumors in children. Available therapeutic agents are not highly effective for treatment of malignant neuroblastomas. Therefore, new treatment strategies are urgently needed. We tested the efficacy of combination of SU5416 (SU), an inhibitor of the vascular endothelial growth factor receptor-2 (VEGFR-2), and (-)-epigallocatechin-3-gallate (EGCG), a polyphenolic compound from green tea, for controlling growth of human malignant neuroblastoma SH-SY5Y and SK-N-BE2 cells. Combination of 20 μM SU and 50 μM EGCG synergistically inhibited cell survival, suppressed expression of VEGFR-2, inhibited cell migration, caused cell cycle arrest, and induced apoptosis. Combination of SU and EGCG effectively blocked angiogenic and survival pathways and modulated expression of cell cycle regulators. Apoptosis was induced by down regulation of Bcl-2, activation of caspase-3, and cleavage of the DNA repair enzyme poly(ADP-ribose) polymerase (PARP). Taken together, this combination of drugs can be a promising therapeutic strategy for controlling the growth of human malignant neuroblastoma cells.
Collapse
Affiliation(s)
- Nishant Mohan
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | | | | | | |
Collapse
|
15
|
|
16
|
Aranha AMF, Zhang Z, Neiva KG, Costa CAS, Hebling J, Nör JE. Hypoxia enhances the angiogenic potential of human dental pulp cells. J Endod 2010; 36:1633-7. [PMID: 20850667 DOI: 10.1016/j.joen.2010.05.013] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 05/25/2010] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Trauma can result in the severing of the dental pulp vessels, leading to hypoxia and ultimately to pulp necrosis. Improved understanding of mechanisms underlying the response of dental pulp cells to hypoxic conditions might lead to better therapeutic alternatives for patients with dental trauma. The purpose of this study was to evaluate the effect of hypoxia on the angiogenic response mediated by human dental pulp stem cells (DPSCs) and human dental pulp fibroblasts (HDPFs). METHODS DPSCs and HDPFs were exposed to experimental hypoxic conditions. Hypoxia-inducible transcription factor-1alpha (HIF-1alpha) was evaluated by Western blot and immunocytochemistry, whereas vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) expression was evaluated by enzyme-linked immunosorbent assay. YC-1, an inhibitor of HIF-1alpha, was used to evaluate the functional effect of this transcriptional factor on hypoxia-induced VEGF expression. Conditioned medium from hypoxic and normoxic pulp cells was used to stimulate human dermal microvascular endothelial cells (HDMECs). HDMEC proliferation was measured by WST-1 assay, and angiogenic potential was evaluated by a capillary sprouting assay in 3-dimensional collagen matrices. RESULTS Hypoxia enhanced HIF-1alpha and VEGF expression in DPSCs and HDPFs. In contrast, hypoxia did not induce bFGF expression in pulp cells. YC-1 partially inhibited hypoxia-induced HIF-1alpha and VEGF in these cells. The growth factor milieu of hypoxic HDPFs (but not hypoxic DPSCs) induced endothelial cell proliferation and sprouting as compared with medium from normoxic cells. CONCLUSIONS Collectively, these data demonstrate that hypoxia induces complex and cell type-specific pro-angiogenic responses and suggest that VEGF (but not bFGF) participates in the revascularization of hypoxic dental pulps.
Collapse
Affiliation(s)
- Andreza M F Aranha
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
Cancers use a nanoscale messenger system known as exosomes to communicate with surrounding tissues and immune cells. However, the functional relationship between tumor exosomes, endothelial signaling, angiogenesis, and metastasis is poorly understood. Herein, we describe a standardized approach for defining the angiogenic potential of isolated exosomes. We created a powerful technique to rapidly and efficiently isolate and track exosomes for study using dynamic light scattering in conjunction with fluorescent exosome labeling. With these methods, melanoma exosomes were observed to interact with and influence endothelial tubule morphology as well as move between endothelial tubule cells by means of tunneling nanotube structures. Melanoma exosomes also were observed to rapidly stimulate the production of endothelial spheroids and endothelial sprouts in a dose-dependent manner. In concert, tumor exosomes simultaneously elicited paracrine endothelial signaling by regulation of certain inflammatory cytokines. These data suggest that, tumor exosomes can promote endothelial angiogenic responses, which could contribute to tumor metastatic potential.
Collapse
|
18
|
Steffens L, Wenger A, Stark GB, Finkenzeller G. In vivo engineering of a human vasculature for bone tissue engineering applications. J Cell Mol Med 2009; 13:3380-6. [PMID: 18624770 PMCID: PMC4516493 DOI: 10.1111/j.1582-4934.2008.00418.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The neovascularization of three-dimensional voluminous tissues, such as bone, represents an important challenge in tissue engineering applications. The formation of a preformed vascular plexus could maintain cell viability and promote vascularization after transplantation. We have developed a three-dimensional spheroidal coculture system consisting of human primary endothelial cells and human primary osteoblasts (hOBs) to improve angiogenesis in bone tissue engineering applications. In this study, we investigated the survival and vascularization of the engineered implants in vivo. Endothelial cell spheroids were cocultured with hOBs in fibrin and seeded into scaffolds consisting of processed bovine cancellous bone (PBCB). The cell-seeded scaffolds were evaluated for their angiogenic potential in two different in vivo assays: the chick embryo chorioallantoic membrane (CAM) model and the severe combined immunodeficiency disorder (SCID) mouse model. In both assays, the development of a complex three-dimensional network of perfused human neovessels could be detected. After subcutaneous implantation into immunodeficient mice, the newly formed human vasculature was stabilized by the recruitment of murine smooth muscle α-actin-positive mural cells and anastomoses with the mouse vasculature. We conclude that this endothelial cell spheroid system can be used to create a network of functional perfused blood vessels in vivo. The finding that this process takes place with high efficacy in the presence of co-implanted primary osteoblasts and in an osteoconductive environment provided by the PBCB scaffold, suggests that this system may be suitable for improving vascularization in bone tissue engineering.
Collapse
Affiliation(s)
- Lilian Steffens
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | |
Collapse
|
19
|
Finkenzeller G, Torio-Padron N, Momeni A, Mehlhorn AT, Stark GB. In vitro angiogenesis properties of endothelial progenitor cells: a promising tool for vascularization of ex vivo engineered tissues. ACTA ACUST UNITED AC 2007; 13:1413-20. [PMID: 17550338 DOI: 10.1089/ten.2006.0369] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Survival of ex vivo constructed tissues after transplantation is limited by insufficient oxygen and nutrient supply. Therefore, strategies aiming at the improvement of neovascularization of engineered tissues are a key issue. A method to enhance graft vascularization is to establish a primitive vascular plexus within the graft before transplantation by the use of cellular-based concepts. To explore the utility of endothelial progenitor cells (EPCs) for the ex vivo vascularization of tissue engineered grafts, we analyzed the in vitro angiogenic properties of this cell type in two different angiogenesis models: the 3-dimensional spheroid sprouting assay and the 2-dimensional matrigel assay. In both assays, EPCs were able to form tubelike structures, resembling early capillaries. This process was significantly enhanced by the addition of angiogenic growth factors. Direct comparison between EPCs and mature endothelial cells, represented by human umbilical vein endothelial cells (HUVECs), revealed that both cell types displayed an almost identical angiogenic potential. Other functional in vitro parameters such as angiogenic growth factor induced cell proliferation and cell survival were investigated as well, revealing a significantly decreased level of apoptosis of EPCs in relation to HUVECs under serum-deprived conditions. The observed survival advantage of EPCs along with the observation that EPCs perform very well in the above mentioned in vitro angiogenesis assays, make them an ideal autologous cell source for vascularization of ex vivo generated tissues. The attractiveness of this cell type for tissue engineering applications is strengthened further by the fact that these cells can be easily isolated from the peripheral blood of patients, thereby eliminating donor site morbidity.
Collapse
Affiliation(s)
- Günter Finkenzeller
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Hugstetter Str. 55, D-79106 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
20
|
Klöpper J, Lindenmaier W, Fiedler U, Mehlhorn A, Stark GB, Finkenzeller G. High efficient adenoviral-mediated VEGF and Ang-1 gene delivery into osteogenically differentiated human mesenchymal stem cells. Microvasc Res 2007; 75:83-90. [PMID: 17603084 DOI: 10.1016/j.mvr.2007.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 04/13/2007] [Accepted: 04/25/2007] [Indexed: 11/16/2022]
Abstract
Survival of ex vivo constructed tissues after transplantation is limited by insufficient oxygen and nutrient supply. Therefore, strategies aiming at improvement of neovascularization of engineered tissues are a key issue in tissue engineering applications. This in vitro study aimed at exploring the usability of osteogenically differentiated human mesenchymal stem cells (MSCs) as carriers of the angiogenic growth factor genes vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) for therapeutic angiogenesis in bone tissue engineering. The ex vivo adenoviral vector mediated transduction into osteogenically differentiated MSCs revealed a highly efficient and long lasting expression of the transgenes. Biological activity of VEGF and Ang-1 secreted from transduced cells was confirmed by analyzing the sprouting, proliferation and apoptosis of human umbilical vein endothelial cells (HUVECs) in response to conditioned medium obtained from transduced cells. The transduced osteogenically differentiated MSCs described in this report may be suitable for inducing neovascularization in bone tissue engineering applications.
Collapse
Affiliation(s)
- Jonas Klöpper
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Amura CR, Brodsky KS, Groff R, Gattone VH, Voelkel NF, Doctor RB. VEGF receptor inhibition blocks liver cyst growth in pkd2(WS25/-) mice. Am J Physiol Cell Physiol 2007; 293:C419-28. [PMID: 17475663 DOI: 10.1152/ajpcell.00038.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proliferation of cyst-lining epithelial cells is an integral part of autosomal dominant polycystic kidney disease (ADPKD) cyst growth. Cytokines and growth factors within cyst fluids are positioned to induce cyst growth. Vascular endothelial growth factor (VEGF) is a pleiotropic growth factor present in ADPKD liver cyst fluids (human 1,128 +/- 78, mouse 2,787 +/- 136 pg/ml) and, to a lesser extent, in ADPKD renal cyst fluids (human 294 +/- 41, mouse 191 +/- 90 pg/ml). Western blotting showed that receptors for VEGF (VEGFR1 and VEGFR2) were present in both normal mouse bile ducts and pkd2(WS25/-) liver cyst epithelial cells. Treatment of pkd2(WS25/-) liver cyst epithelial cells with VEGF (50-50,000 pg/ml) or liver cyst fluid induced a proliferative response. The effect on proliferation of liver cyst fluid was inhibited by SU-5416, a potent VEGF receptor inhibitor. Treatment of pkd2(WS25/-) mice between 4 and 8 mo of age with SU-5416 markedly reduced the cyst volume density of the liver (vehicle 9.9 +/- 4.3%, SU-5416 1.8 +/- 0.7% of liver). SU-5416 treatment between 4 and 12 mo of age markedly protected against increases in liver weight [pkd2(+/+) 4.8 +/- 0.2%, pkd2(WS25/-)-vehicle 10.8 +/- 1.9%, pkd2(WS25/-)-SU-5416 4.8 +/- 0.4% body wt]. The capacity of VEGF signaling to induce in vitro proliferation of pkd2(WS25/-) liver cyst epithelial cells and inhibition of in vivo VEGF signaling to retard liver cyst growth in pkd2(WS25/-) mice indicates that the VEGF signaling pathway is a potentially important therapeutic target in the treatment of ADPKD liver cyst disease.
Collapse
MESH Headings
- Animals
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyst Fluid/metabolism
- Cysts/genetics
- Cysts/metabolism
- Cysts/pathology
- Cysts/prevention & control
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Indoles/pharmacology
- Indoles/therapeutic use
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver Diseases/genetics
- Liver Diseases/metabolism
- Liver Diseases/pathology
- Liver Diseases/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Polycystic Kidney, Autosomal Dominant/complications
- Polycystic Kidney, Autosomal Dominant/drug therapy
- Polycystic Kidney, Autosomal Dominant/genetics
- Polycystic Kidney, Autosomal Dominant/metabolism
- Polycystic Kidney, Autosomal Dominant/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyrroles/pharmacology
- Pyrroles/therapeutic use
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Signal Transduction/drug effects
- TRPP Cation Channels/metabolism
- Time Factors
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/pharmacology
- Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-1/metabolism
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Claudia R Amura
- Department of Medicine, University of Colorado Health Sciences Center, Denver, USA
| | | | | | | | | | | |
Collapse
|
22
|
Li Y, He L. Establishment of the model of vascular endothelial cell membrane chromatography and its preliminary application. ACTA ACUST UNITED AC 2007. [DOI: 10.1007/s11434-007-0149-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Yance DR, Sagar SM. Targeting angiogenesis with integrative cancer therapies. Integr Cancer Ther 2006; 5:9-29. [PMID: 16484711 DOI: 10.1177/1534735405285562] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An integrative approach for managing a patient with cancer should target the multiple biochemical and physiological pathways that support tumor development while minimizing normal tissue toxicity. Angiogenesis is a key process in the promotion of cancer. Many natural health products that inhibit angiogenesis also manifest other anticancer activities. The authors will focus on natural health products (NHPs) that have a high degree of antiangiogenic activity but also describe some of their many other interactions that can inhibit tumor progression and reduce the risk of metastasis. NHPs target various molecular pathways besides angiogenesis, including epidermal growth factor receptor (EGFR), the HER-2/neu gene, the cyclooxygenase-2 enzyme, the NF-kB transcription factor, the protein kinases, Bcl-2 protein, and coagulation pathways. The herbalist has access to hundreds of years of observational data on the anticancer activity of many herbs. Laboratory studies are confirming the knowledge that is already documented in traditional texts. The following herbs are traditionally used for anticancer treatment and are antiangiogenic through multiple interdependent processes that include effects on gene expression, signal processing, and enzyme activities: Artemisia annua (Chinese wormwood), Viscum album (European mistletoe), Curcuma longa (turmeric), Scutellaria baicalensis (Chinese skullcap), resveratrol and proanthocyanidin (grape seed extract), Magnolia officinalis (Chinese magnolia tree), Camellia sinensis (green tea), Ginkgo biloba, quercetin, Poria cocos, Zingiber officinale (ginger), Panax ginseng, Rabdosia rubescens (rabdosia), and Chinese destagnation herbs. Quality assurance of appropriate extracts is essential prior to embarking on clinical trials. More data are required on dose response, appropriate combinations, and potential toxicities. Given the multiple effects of these agents, their future use for cancer therapy probably lies in synergistic combinations. During active cancer therapy, they should generally be evaluated in combination with chemotherapy and radiation. In this role, they act as biological response modifiers and adaptogens, potentially enhancing the efficacy of the so-called conventional therapies. Their effectiveness may be increased when multiple agents are used in optimal combinations. New designs for trials to demonstrate activity in human subjects are required. Although controlled trials might be preferred, smaller studies with appropriate end points and surrogate markers for antiangiogenic response could help prioritize agents for the larger resource-intensive phase 3 trials.
Collapse
|
24
|
Wenger A, Kowalewski N, Stahl A, Mehlhorn AT, Schmal H, Stark GB, Finkenzeller G. Development and characterization of a spheroidal coculture model of endothelial cells and fibroblasts for improving angiogenesis in tissue engineering. Cells Tissues Organs 2006; 181:80-8. [PMID: 16534202 DOI: 10.1159/000091097] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Indexed: 01/29/2023] Open
Abstract
Neovascularization is a critical step in tissue engineering applications since implantation of voluminous grafts without sufficient vascularity results in hypoxic cell death of central tissues. We have developed a three-dimensional spheroidal coculture system consisting of human umbilical vein endothelial cells (HUVECs) and human primary fibroblasts (hFBs) to improve angiogenesis in tissue engineering applications. Morphological analysis of cryosections from HUVEC/hFB cospheroids revealed a characteristic temporal and spatial organization with HUVECs located in the center of the cospheroid and a peripheral localization of fibroblasts. In coculture spheroids, the level of apoptosis of endothelial cells was strongly decreased upon cocultivation with fibroblasts. Collagen-embedded HUVEC spheroids develop numerous lumenized capillary-like sprouts. This was also apparent for HUVEC/hFB cospheroids, albeit to a lesser extent. Quantification of cumulative sprout length revealed an approximately 35% reduction in endothelial cell sprouting upon cocultivation with fibroblasts in cospheroids. The slight reduction in endothelial cell sprouting was not mediated by a paracrine mechanism but is most likely due to the formation of heterogenic cell contacts between HUVECs and hFBs within the cospheroid. The model system introduced in this study is suitable for the development of a preformed lumenized capillary-like network ex vivo and may therefore be useful for improving angiogenesis in in vivo tissue engineering applications.
Collapse
Affiliation(s)
- Andreas Wenger
- Department of Plastic, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Abadi AH, Abou-Seri SM, Abdel-Rahman DE, Klein C, Lozach O, Meijer L. Synthesis of 3-substituted-2-oxoindole analogues and their evaluation as kinase inhibitors, anticancer and antiangiogenic agents. Eur J Med Chem 2006; 41:296-305. [PMID: 16494969 DOI: 10.1016/j.ejmech.2005.12.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 11/17/2005] [Accepted: 12/05/2005] [Indexed: 10/25/2022]
Abstract
Several analogues of the 3-substituted-2-oxoindole chemotype were synthesized by condensing isatin or the appropriate haloisatin with some amino acids or histamine under neutral conditions. All the imino derivatives produced were tested for kinase inhibitory properties against three serine/threonine kinases, namely CDK1/cyclin B, CDK5/p25 and GSK3alpha/beta. Most of the histidine derivatives showed inhibitory properties to the three kinases in the low micromolar range. The histamine derivatives were less potent against CDK1/cyclin B and CDK5/p25 and totally inactive against GSK3alpha/beta. So, the management of the carboxyl function may be a tool to impart selectivity in such family of kinases. Docking of 2-[[-5-bromo-2-oxoindolin-3-ylidene]amino]-3-(1H-imidazol2-yl)propanoic acid 14 to CDK5/p25 indicates that this compound can interact with the enzyme through four hydrogen bonds; for GSK/3beta, the ligand poses itself in another orientation, also four hydrogen bonds can be formed between the ligand and the receptor, otherwise hydrophobic interactions seem to predominate. Also, all the final compounds were tested for their in vitro antitumor properties against MCF7 (breast), NCI-H460 (lung) and SF268 (CNS) cancer cell lines. None of the synthesized compounds was cytotoxic at 10(-4) molar concentration. Moreover, compounds 13 and 14 were tested for potential antiangiogenic properties by testing their ability to inhibit the proliferation of human umbilical vein endothelial cells (HUVECs), cord formation and migration in response to chemoattractant. Only compound 14 showed moderate inhibitory properties to HUVECs proliferation and cord formation while its non-brominated derivative 13 did not. Thus, the antiangiogenesis properties are not apparently caused by inhibition of any of the tested kinases.
Collapse
Affiliation(s)
- Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Egypt.
| | | | | | | | | | | |
Collapse
|
26
|
Bargagna-Mohan P, Baek SH, Lee H, Kim K, Mohan R. Use of PROTACS as molecular probes of angiogenesis. Bioorg Med Chem Lett 2005; 15:2724-7. [PMID: 15876533 PMCID: PMC3226786 DOI: 10.1016/j.bmcl.2005.04.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 03/30/2005] [Accepted: 04/04/2005] [Indexed: 10/25/2022]
Abstract
Small molecules designed to specifically activate or inactivate protein functions have been useful to study biological processes. PROTACS are small molecule chimera which comprise a ligand and a peptide recognition motif for an E3 ligase. These novel reagents exploit the ubiquitin-mediated proteasome degradation pathway to target the ligand-bound protein for intracellular degradation. Here, we report that an estrogen receptor (ER)-targeting PROTACS that causes degradation of ER is able to potently inhibit endothelial cell differentiation in a three-dimensional angiogenic sprouting assay. These findings support the use of ER-targeting PROTACS as probes of angiogenesis.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Sun-Hee Baek
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Hyosung Lee
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kyungbo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Royce Mohan
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
27
|
Peterson AC, Swiger S, Stadler WM, Medved M, Karczmar G, Gajewski TF. Phase II study of the Flk-1 tyrosine kinase inhibitor SU5416 in advanced melanoma. Clin Cancer Res 2005; 10:4048-54. [PMID: 15217937 DOI: 10.1158/1078-0432.ccr-03-0766] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF) expression is prognostic in melanoma, and the activity of VEGF is mediated in part through the receptor tyrosine kinase Flk-1. A Phase II study of SU5416, a preferential inhibitor of Flk-1, was carried out in patients with metastatic melanoma to determine clinical response, tolerability, and changes in tumor vascular perfusion. EXPERIMENTAL DESIGN Patients with documented progressive disease and </=1 prior therapy were eligible. Central nervous system metastases were allowed if stable off medication. SU5416 (145 mg/m(2)) was administered via a central catheter twice weekly for 8 weeks. Premedication with dexamethasone, diphenhydramine, and a H(2) blocker was required because of the Cremophor vehicle. Tumor vascular perfusion was assessed before treatment and during week 8 by dynamic contrast magnetic resonance imaging, and plasma was analyzed for VEGF. RESULTS Thirty-one patients were enrolled. Two-thirds had received prior therapy, 21 had visceral metastasis, and 14 had an elevated lactate dehydrogenase. Mean absolute lymphocyte counts were decreased (P = 0.002), and glucose levels were increased (P = 0.001) posttherapy, presumably because of steroid premedication. Four vascular adverse events were observed. Of 26 evaluable patients, 1 experienced a partial response, 1 had stable disease, and 5 had a mixed response. Dynamic contrast magnetic resonance imaging in 5 evaluable patients showed decreased tumor perfusion at week 8 (P = 0.024), and plasma VEGF levels were elevated compared with pretherapy (P = 0.008). CONCLUSIONS SU5146 appears to be relatively well tolerated in this population. Although the modest clinical activity and potential effects on tumor vascularity may support additional exploration of VEGF as a target in melanoma, effects from steroid premedication limit further investigation of this agent.
Collapse
Affiliation(s)
- Amy C Peterson
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Cancer Research Center, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
28
|
Stewart DJ, Edwards JG, Smythe WR, Waller DA, O'Byrne KJ. Malignant pleural mesothelioma--an update. INTERNATIONAL JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL HEALTH 2004; 10:26-39. [PMID: 15070023 DOI: 10.1179/oeh.2004.10.1.26] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Exposure to asbestos is the most frequent, but not exclusive, cause of malignant mesothelioma. Clinical features include dyspnea, cough, nonspecific chest pain, weight loss and night sweats. Diagnosis may be complicated by histologic difficulties. Thoracoscopic techniques are proving beneficial, but no one method of imaging has proven superior, and disease staging is inconsistent. Conventional treatments such as chemotherapy, surgery, and radiotherapy have had variable impacts, although chemotherapy is useful in palliation and can improve both survival and quality of life. There is hope for new antimetabolite agents. The role of radical surgery is yet to be evaluated in a large trial. New radiotherapeutic techniques to improve local control are promising. Multimodality treatments appear to be the most successful for management of potentially resectable disease. It is likely that biological markers will improve accuracy in staging and prognosis. With new treatments based on better understanding of the biology of the disease, there is cautious optimism for the future for patients with malignant pleural mesothelioma.
Collapse
Affiliation(s)
- Duncan J Stewart
- University Department of Oncology, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | | | | | | | | |
Collapse
|
29
|
Timmins NE, Dietmair S, Nielsen LK. Hanging-drop multicellular spheroids as a model of tumour angiogenesis. Angiogenesis 2004; 7:97-103. [PMID: 15516830 DOI: 10.1007/s10456-004-8911-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The establishment of a vascular network within tumours is a key step in the progression towards an aggressive, metastatic state, with poor prognosis. We have developed a novel in vitro model to specifically capture the interaction between endothelial cells and solid tumours. Micro-vascularised in vitro tumour constructs were produced by introducing endothelial cells to multicellular spheroids formed in hanging drops. Upon introduction, the endothelial cells migrated into the tumour spheroid, establishing tubular networks and luminal structures. This system relies on the natural pro-angiogenic capacity of multicellular spheroids, and does not require the addition of exogenous angiogenic factors, or use of extracellular-matrix substitutes.
Collapse
Affiliation(s)
- Nicholas E Timmins
- Laboratory for Biological Engineering, Department of Chemical Engineering, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | | | | |
Collapse
|
30
|
Chen JX, Lawrence ML, Cunningham G, Christman BW, Meyrick B. HSP90 and Akt modulate Ang-1-induced angiogenesis via NO in coronary artery endothelium. J Appl Physiol (1985) 2003; 96:612-20. [PMID: 14555685 DOI: 10.1152/japplphysiol.00728.2003] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study examines the notion that heat shock protein (HSP) 90 binding to nitric oxide (NO), endothelial NO synthase (eNOS), and PI3K-Akt regulate angiopoietin (Ang)-1-induced angiogenesis in porcine coronary artery endothelial cells (PCAEC). Exposure to Ang-1 (250 ng/ml) for periods up to 2 h resulted in a time-dependent increase in eNOS phosphorylation at Ser 1177 that occurred by 5 min and peaked at 60 min. This was accompanied by a gradual increase in NO release. Ang-1 also led to stimulation of HSP90 binding to eNOS and a significant increase in Akt phosphorylation. Thirty minutes of pretreatment of cells with either 1 microg/ml geldanamycin (a specific inhibitor of HSP90) or 500 nM wortmannin [a specific phosphatidylinositol 3 (PI3)-kinase (PI3K) inhibitor] significantly attenuated Ang-1-stimulated eNOS phosphorylation and NO production. Exposure to Ang-1 caused an increase in endothelial cell migration, tube formation, and sprouting from PCAEC spheroids, and pharmacological blockage of HSP90 function or inhibition of PI3K-Akt pathway completely abolished these effects. Inhibition of nitric oxide synthase by NG-nitro-l-arginine methyl ester (2.5 mM) also resulted in a significant decrease in Ang-1-induced angiogenesis. We conclude that stimulated HSP90 binding to eNOS and activation of the PI3-Akt pathway contribute to Ang-1-induced eNOS phosphorylation, NO production, and angiogenesis in PCAEC.
Collapse
Affiliation(s)
- Jian-xiong Chen
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2650, USA
| | | | | | | | | |
Collapse
|