1
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Antiaging Strategies and Remedies: A Landscape of Research Progress and Promise. ACS Chem Neurosci 2024; 15:408-446. [PMID: 38214973 PMCID: PMC10853939 DOI: 10.1021/acschemneuro.3c00532] [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: 08/12/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Aging is typified by a gradual loss of physiological fitness and accumulation of cellular damage, leading to deteriorated functions and enhanced vulnerability to diseases. Antiaging research has a long history throughout civilization, with many efforts put forth to understand and prevent the effects of aging. Multiple strategies aiming to promote healthy aging and extend the lifespan have been developed including lifestyle adjustments, medical treatments, and social programs. A multitude of antiaging medicines and remedies have also been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent research related to antiaging strategies and treatments. We review the recent advances and delineate trends in research headway of antiaging knowledge and practice across time, geography, and development pipelines. We further assess the state-of-the-art antiaging approaches and explore their correlations with age-related diseases. The landscape of antiaging drugs has been outlined and explored. Well-recognized and novel, currently evaluated antiaging agents have also been summarized. Finally, we review clinical applications of antiaging products with their development pipelines. The objective of this review is to summarize current knowledge on preventive strategies and treatment remedies in the field of aging, to outline challenges and evaluate growth opportunities, in order to further efforts to solve the problems that remain.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
2
|
Esparza A, Jimenez N, Borrego EA, Browne S, Natividad-Diaz SL. Review: Human stem cell-based 3D in vitro angiogenesis models for preclinical drug screening applications. Mol Biol Rep 2024; 51:260. [PMID: 38302762 PMCID: PMC10834608 DOI: 10.1007/s11033-023-09048-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 11/06/2023] [Indexed: 02/03/2024]
Abstract
Vascular diseases are the underlying pathology in many life-threatening illnesses. Human cellular and molecular mechanisms involved in angiogenesis are complex and difficult to study in current 2D in vitro and in vivo animal models. Engineered 3D in vitro models that incorporate human pluripotent stem cell (hPSC) derived endothelial cells (ECs) and supportive biomaterials within a dynamic microfluidic platform provide a less expensive, more controlled, and reproducible platform to better study angiogenic processes in response to external chemical or physical stimulus. Current studies to develop 3D in vitro angiogenesis models aim to establish single-source systems by incorporating hPSC-ECs into biomimetic extracellular matrices (ECM) and microfluidic devices to create a patient-specific, physiologically relevant platform that facilitates preclinical study of endothelial cell-ECM interactions, vascular disease pathology, and drug treatment pharmacokinetics. This review provides a detailed description of the current methods used for the directed differentiation of human stem cells to endothelial cells and their use in engineered 3D in vitro angiogenesis models that have been developed within the last 10 years.
Collapse
Affiliation(s)
- Aibhlin Esparza
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Nicole Jimenez
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Edgar A Borrego
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Shane Browne
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA.
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
3
|
Esparza A, Jimenez N, Joddar B, Natividad-Diaz S. Development of in vitro cardiovascular tissue models within capillary circuit microfluidic devices fabricated with 3D Stereolithography printing. RESEARCH SQUARE 2023:rs.3.rs-2667200. [PMID: 36993455 PMCID: PMC10055652 DOI: 10.21203/rs.3.rs-2667200/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human cardiovascular tissue and diseases are difficult to study for novel drug discovery and fundamental cellular/molecular processes due to limited availability of physiologically-relevant models in vitro.[1-3] Animal models may resemble human heart structure, however there are significant differences from human cardiovascular physiology including biochemical signaling, and gene expression.[4-6] In vitro microfluidic tissue models provide a less expensive, more controlled, and reproducible platform for better quantification of isolated cellular processes in response to biochemical or biophysical stimulus.[6-12] The capillary driven-flow microfluidic device in this study was manufactured with a 3D stereolithography (SLA) printed mold and is a closed circuit system operating on principles of capillary action allowing continuous fluid movement without external power supply. Human umbilical vein endothelial cells (HUVECs) and human cardiomyocytes (AC16) were encapsulated into a fibrin hydrogel to form vascular (VTM) and cardiac (CTM) tissue models respectively. To determine response to biophysical stimulus, the 3D cardiovascular tissue was directly loaded into the device tissue culture chambers that either had no microposts (DWoP) or microposts (DWPG) for 1, 3 and 5 days. The tissues were analyzed with fluorescent microscopy for morphological differences, average tube length, and cell orientation between tissues cultured in both conditions. In DWPG VTMs displayed capillary-like tube formation with visible cell alignment and orientation, while AC16s continued to elongate around microposts by day 5. VTM and CTM models in devices with posts (DWPG) displayed cell alignment and orientation after 5 days, indicated the microposts induced biophysical cues to guide cell structure and specific organization.
Collapse
|
4
|
Hann SY, Cui H, Chen G, Boehm M, Esworthy T, Zhang LG. 3D printed biomimetic flexible blood vessels with iPS cell-laden hierarchical multilayers. BIOMEDICAL ENGINEERING ADVANCES 2022; 4:100065. [PMID: 36582411 PMCID: PMC9794201 DOI: 10.1016/j.bea.2022.100065] [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] [Indexed: 11/29/2022] Open
Abstract
Successful recovery from vascular diseases has typically relied on the surgical repair of damaged blood vessels (BVs), with the majority of current approaches involving the implantation of autologous BVs, which is plagued by donor site tissue damage. Researchers have attempted to develop artificial vessels as an alternative solution to traditional approaches to BV repair. However, the manufacturing of small-diameter (< 6 mm) BVs is still considered one of the biggest challenges due to its difficulty in the precise fabrication and the replication of biomimetic architectures. In this study, we successfully developed 3D printed flexible small-diameter BVs that consist of smooth muscle cells and a vascularized endothelium. In the developed artificial BV, a rubber-like elastomer was printed as the outermost layer of the vessel, which demonstrated enhanced mechanical properties, while and human induced pluripotent stem cell (iPSC)-derived vascular smooth muscle cells (iSMCs) and endothelial cells (iECs) embedded fibrinogen solutions were coaxially extruded with thrombin solution to form cell-laden fibrin gel inner layers. Our results showed that the 3D BVs possessed proper mechanical properties, and the cells in the fibrin layers substantially proliferated over time to form a stable BV construct. Our study demonstrated that the 3D printed flexible small-diameter BV using iPSCs could be a promising platform for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Guibin Chen
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Medicine, The George Washington University Medical Center, Washington, DC 20052, USA
| |
Collapse
|
5
|
A new hemostatic agent composed of Zn2+-enriched Ca2+ alginate activates vascular endothelial cells in vitro and promotes tissue repair in vivo. Bioact Mater 2022; 18:368-382. [PMID: 35415309 PMCID: PMC8965972 DOI: 10.1016/j.bioactmat.2022.01.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/28/2022] Open
Abstract
To control capillary bleeding, surgeons may use absorbable hemostatic agents, such as Surgicel® and TachoSil®. Due to their slow resorption, their persistence in situ can have a negative impact on tissue repair in the resected organ. To avoid complications and obtain a hemostatic agent that promotes tissue repair, a zinc-supplemented calcium alginate compress was developed: HEMO-IONIC®. This compress is non-absorbable and is therefore removed once hemostasis has been achieved. After demonstrating the hemostatic efficacy and stability of the blood clot obtained with HEMO-IONIC, the impact of Surgicel, TachoSil, and HEMO-IONIC on cell activation and tissue repair were compared (i) in vitro on endothelial cells, which are essential to tissue repair, and (ii) in vivo in a mouse skin excision model. In vitro, only HEMO-IONIC maintained the phenotypic and functional properties of endothelial cells and induced their migration. In comparison, Surgicel was found to be highly cytotoxic, and TachoSil inhibited endothelial cell migration. In vivo, only HEMO-IONIC increased angiogenesis, the recruitment of cells essential to tissue repair (macrophages, fibroblasts, and epithelial cells), and accelerated maturation of the extracellular matrix. These results demonstrate that a zinc-supplemented calcium alginate, HEMO-IONIC, applied for 10 min at the end of surgery and then removed has a long-term positive effect on all phases of tissue repair. A new Zn2+ enriched Ca2+ alginate hemostatic agent, HEMO-IONIC, has been developed. Non-absorbable, it achieves hemostasis with no foreign bodies left in the wound. HEMO-IONIC stimulates endothelial cell migration in vitro and angiogenesis in vivo. HEMO-IONIC, removed 10 min after application, promotes all stages of tissue repair.
Collapse
|
6
|
Joddar B, Natividad-Diaz SL, Padilla AE, Esparza AA, Ramirez SP, Chambers DR, Ibaroudene H. Engineering approaches for cardiac organoid formation and their characterization. Transl Res 2022; 250:46-67. [PMID: 35995380 PMCID: PMC10370285 DOI: 10.1016/j.trsl.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
Cardiac organoids are 3-dimensional (3D) structures composed of tissue or niche-specific cells, obtained from diverse sources, encapsulated in either a naturally derived or synthetic, extracellular matrix scaffold, and include exogenous biochemical signals such as essential growth factors. The overarching goal of developing cardiac organoid models is to establish a functional integration of cardiomyocytes with physiologically relevant cells, tissues, and structures like capillary-like networks composed of endothelial cells. These organoids used to model human heart anatomy, physiology, and disease pathologies in vitro have the potential to solve many issues related to cardiovascular drug discovery and fundamental research. The advent of patient-specific human-induced pluripotent stem cell-derived cardiovascular cells provide a unique, single-source approach to study the complex process of cardiovascular disease progression through organoid formation and incorporation into relevant, controlled microenvironments such as microfluidic devices. Strategies that aim to accomplish such a feat include microfluidic technology-based approaches, microphysiological systems, microwells, microarray-based platforms, 3D bioprinted models, and electrospun fiber mat-based scaffolds. This article discusses the engineering or technology-driven practices for making cardiac organoid models in comparison with self-assembled or scaffold-free methods to generate organoids. We further discuss emerging strategies for characterization of the bio-assembled cardiac organoids including electrophysiology and machine-learning and conclude with prospective points of interest for engineering cardiac tissues in vitro.
Collapse
Affiliation(s)
- Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Andie E Padilla
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Aibhlin A Esparza
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | | | | |
Collapse
|
7
|
Functional Characterization of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Int J Mol Sci 2022; 23:ijms23158507. [PMID: 35955642 PMCID: PMC9368986 DOI: 10.3390/ijms23158507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/03/2022] Open
Abstract
Endothelial cells derived from human induced pluripotent stem cells (hiPSC-ECs) provide a new opportunity for mechanistic research on vascular regeneration and drug screening. However, functions of hiPSC-ECs still need to be characterized. The objective of this study was to investigate electrophysiological and functional properties of hiPSC-ECs compared with primary human cardiac microvascular endothelial cells (HCMECs), mainly focusing on ion channels and membrane receptor signaling, as well as specific cell functions. HiPSC-ECs were derived from hiPS cells that were generated from human skin fibroblasts of three independent healthy donors. Phenotypic and functional comparison to HCMECs was performed by flow cytometry, immunofluorescence staining, quantitative reverse-transcription polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA), tube formation, LDL uptake, exosome release assays and, importantly, patch clamp techniques. HiPSC-ECs were successfully generated from hiPS cells and were identified by endothelial markers. The mRNA levels of KCNN2, KCNN4, KCNMA1, TRPV2, and SLC8A1 in hiPSC-ECs were significantly higher than HCMECs. AT1 receptor mRNA level in hiPSC-ECs was higher than in HCMECs. AT2 receptor mRNA level was the highest among all receptors. Adrenoceptor ADRA2 expression in hiPSC-ECs was lower than in HCMECs, while ADRA1, ADRB1, ADRB2, and G-protein GNA11 and Gai expression were similar in both cell types. The expression level of muscarinic and dopamine receptors CHRM3, DRD2, DRD3, and DRD4 in hiPSC-ECs were significantly lower than in HCMECs. The functional characteristics of endothelial cells, such as tube formation and LDL uptake assay, were not statistically different between hiPSC-ECs and HCMECs. Phenylephrine similarly increased the release of the vasoconstrictor endothelin-1 (ET-1) in hiPSC-ECs and HCMECs. Acetylcholine also similarly increased nitric oxide generation in hiPSC-ECs and HCMECs. The resting potentials (RPs), ISK1–3, ISK4 and IK1 were similar in hiPSC-ECs and HCMECs. IBK was larger and IKATP was smaller in hiPSC-ECs. In addition, we also noted a higher expression level of exosomes marker CD81 in hiPSC-ECs and a higher expression of CD9 and CD63 in HCMECs. However, the numbers of exosomes extracted from both types of cells did not differ significantly. The study demonstrates that hiPSC-ECs are similar to native endothelial cells in ion channel function and membrane receptor-coupled signaling and physiological cell functions, although some differences exist. This information may be helpful for research using hiPSC-ECs.
Collapse
|
8
|
Development of vascular disease models to explore disease causation and pathomechanisms of rare vascular diseases. Semin Immunopathol 2022; 44:259-268. [PMID: 35233690 PMCID: PMC8887661 DOI: 10.1007/s00281-022-00925-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
Abstract
As the field of medicine is striving forward heralded by a new era of next-generation sequencing (NGS) and integrated technologies such as bioprinting and biological material development, the utility of rare monogenetic vascular disease modeling in this landscape is starting to emerge. With their genetic simplicity and broader applicability, these patient-specific models are at the forefront of modern personalized medicine. As a collective, rare diseases are a significant burden on global healthcare systems, and rare vascular diseases make up a significant proportion of this. High costs are due to a lengthy diagnostic process, affecting all ages from infants to adults, as well as the severity and chronic nature of the disease. Their complex nature requires sophisticated disease models and integrated approaches involving multidisciplinary teams. Here, we review these emerging vascular disease models, how they contribute to our understanding of the pathomechanisms in rare vascular diseases and provide useful platforms for therapeutic discovery.
Collapse
|
9
|
Hsieh HL, Liang CC, Lu CY, Yang JT, Chung CY, Ko YS, Lee TH. Induced pluripotent stem cells can improve thrombolytic effect of low-dose rt-PA after acute carotid thrombosis in rat. Stem Cell Res Ther 2021; 12:549. [PMID: 34674761 PMCID: PMC8532293 DOI: 10.1186/s13287-021-02615-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022] Open
Abstract
Background Intravenous thrombolysis using recombinant tissue plasminogen activator (rt-PA) is the standard treatment for acute ischemic stroke. Standard-dose rt-PA (0.9 mg/kg) is known to achieve good recanalization but carries a high bleeding risk. Lower dose of rt-PA has less bleeding risk but carries a high re-occlusion rate. We investigate if induced pluripotent stem cells (iPSCs) can improve the thrombolytic effect of low-dose rt-PA (0.45 mg/kg). Methods Single irradiation with 6 mW/cm2 light-emitting diode (LED) for 4 h at rat common carotid artery was used as thrombosis model according to our previous report. Endothelin-1 (ET-1), intercellular adhesion molecule-1 (ICAM-1), and interleukin 1 beta (IL-1 beta) were used as the inflammatory markers for artery endothelial injury. Angiopoietin-2 (AP-2), brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) were examined in artery wall and iPSCs culture. Animal ultrasound was used to evaluate the stenosis degree of common carotid artery before and at 2 h, 24 h, 4 days and 7 days after LED irradiation. Results After LED irradiation alone, there was a persistent occlusion from 2 h to 7 days. Standard-dose rt-PA alone could recanalize the occluded artery from 24 h to 7 days to stenotic degree ≤ 50%. Low-dose rt-PA or 1 × 106 mouse iPSCs alone could not recanalize the occluded arteries from 2 h to 7 days. Combination use of low-dose rt-PA plus 1 × 106 mouse iPSCs caused better recanalization from 24 h to 7 days. ET-1, ICAM-1 and IL-1 beta were strongly expressed after LED irradiation but reduced after iPSCs treatment. AP-2, BDNF and VEGF were rarely induced after LED irradiation but strongly expressed after iPSCs treatment. In vitro study showed iPSCs could express AP-2, BDNF and VEGF. Conclusion The adjuvant use of iPSCs may help improving the thrombolytic effect of low-dose rt-PA by suppressing inflammatory factors and inducing angiogenic trophic factors. Stem cells could be a potential regimen in acute thrombolytic therapy to improve recanalization and reduce complications. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02615-z.
Collapse
Affiliation(s)
- Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Ching-Chung Liang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-You Lu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Shien Ko
- The First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, and College of Medicine, Chang Gung University, No. 5, Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan.
| |
Collapse
|
10
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
11
|
Alle Q, Le Borgne E, Milhavet O, Lemaitre JM. Reprogramming: Emerging Strategies to Rejuvenate Aging Cells and Tissues. Int J Mol Sci 2021; 22:3990. [PMID: 33924362 PMCID: PMC8070588 DOI: 10.3390/ijms22083990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is associated with a progressive and functional decline of all tissues and a striking increase in many "age-related diseases". Although aging has long been considered an inevitable process, strategies to delay and potentially even reverse the aging process have recently been developed. Here, we review emerging rejuvenation strategies that are based on reprogramming toward pluripotency. Some of these approaches may eventually lead to medical applications to improve healthspan and longevity.
Collapse
Affiliation(s)
- Quentin Alle
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| | - Enora Le Borgne
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| | - Ollivier Milhavet
- IRMB, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| |
Collapse
|
12
|
Jeon SB, Seo BG, Baek SK, Lee HG, Shin JH, Lee IW, Kim HJ, Moon SY, Shin KC, Choi JW, Kim TS, Lee JH, Hwangbo C. Endothelial Cells Differentiated from Porcine Epiblast Stem Cells. Cell Reprogram 2021; 23:89-98. [PMID: 33861642 DOI: 10.1089/cell.2020.0088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) have the ability of self-renewal that can retain the characteristics of the mother cell, and of pluripotency that can differentiate into several body types. PSCs typically include embryonic stem cells (ESCs) derived from the inner cell mass of the preimplantation embryo, and epiblast stem cells (EpiSCs) derived from the epiblast of postimplantation embryo. Although PSCs are able to be used by differentiation into endothelial cells as a potential treatment for vascular diseases, human ESCs and induced PSCs (iPSCs) are followed by ethical and safety issues. Pigs are anatomically and physiologically similar to humans. Therefore, the goal of this study was to establish an efficient protocol that differentiates porcine EpiSCs (pEpiSCs) into the endothelial cells for applying the treatment of human vascular diseases. As a result, alkaline phosphatase (AP)-negative (-) pEpiSCs cultured in endothelial cell growth basal medium-2 (EBM-2) differentiation medium in association with 50 ng/mL of vascular endothelial growth factor (VEGF) for 8 days were changed morphologically like the feature of endothelial cells, and expression of pluripotency-associated markers (OCT-3/4, NANOG, SOX2, and C-MYC) in porcine differentiated cells was significantly decreased (p < 0.05). Additionally, when pEpiSCs were cultured in EBM-2 + 50 ng/mL of VEGF, porcine differentiated cells represented a common endothelial cell marker positive (CD31+) but monocytes and lymphocytes marker negative (CD45-). Therefore, these results indicated that pEpiSCs cultured in EBM-2 + 50 ng/mL of VEGF culture condition were efficiently differentiated into endothelial cells for the treatment of blood vessel diseases.
Collapse
Affiliation(s)
- Soo-Been Jeon
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Bo-Gyeong Seo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Geongsang National University, Jinju, Republic of Korea
| | - Sang-Ki Baek
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Hyeon-Geun Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Institute of Agriculture and Life Science, and College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Joon-Hong Shin
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - In-Won Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyo-Jin Kim
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Geongsang National University, Jinju, Republic of Korea
| | - Sun Young Moon
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Geongsang National University, Jinju, Republic of Korea
| | - Keum-Chul Shin
- Institute of Agriculture and Life Science, and College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Department of Forest Environmental Resources, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jung-Woo Choi
- Institute of Agriculture and Life Science, and College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Tae-Suk Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,College of Animal Life Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Cheol Hwangbo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Geongsang National University, Jinju, Republic of Korea
| |
Collapse
|
13
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
14
|
Lu TM, Houghton S, Magdeldin T, Durán JGB, Minotti AP, Snead A, Sproul A, Nguyen DHT, Xiang J, Fine HA, Rosenwaks Z, Studer L, Rafii S, Agalliu D, Redmond D, Lis R. Pluripotent stem cell-derived epithelium misidentified as brain microvascular endothelium requires ETS factors to acquire vascular fate. Proc Natl Acad Sci U S A 2021; 118:e2016950118. [PMID: 33542154 PMCID: PMC7923590 DOI: 10.1073/pnas.2016950118] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells derived from pluripotent sources in vitro must resemble those found in vivo as closely as possible at both transcriptional and functional levels in order to be a useful tool for studying diseases and developing therapeutics. Recently, differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cells (ECs) with blood-brain barrier (BBB)-like properties has been reported. These cells have since been used as a robust in vitro BBB model for drug delivery and mechanistic understanding of neurological diseases. However, the precise cellular identity of these induced brain microvascular endothelial cells (iBMECs) has not been well described. Employing a comprehensive transcriptomic metaanalysis of previously published hPSC-derived cells validated by physiological assays, we demonstrate that iBMECs lack functional attributes of ECs since they are deficient in vascular lineage genes while expressing clusters of genes related to the neuroectodermal epithelial lineage (Epi-iBMEC). Overexpression of key endothelial ETS transcription factors (ETV2, ERG, and FLI1) reprograms Epi-iBMECs into authentic endothelial cells that are congruent with bona fide endothelium at both transcriptomic as well as some functional levels. This approach could eventually be used to develop a robust human BBB model in vitro that resembles the human brain EC in vivo for functional studies and drug discovery.
Collapse
Affiliation(s)
- Tyler M Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Tarig Magdeldin
- Department of Neurology and the Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine-New York Presbyterian Hospital, New York, NY 10065
| | - José Gabriel Barcia Durán
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Andrew P Minotti
- Developmental Biology, the Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- The Biochemistry, Structural Biology, Cell Biology, Developmental Biology and Molecular Biology Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
| | - Amanda Snead
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Andrew Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Duc-Huy T Nguyen
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY 10065
| | - Howard A Fine
- Department of Neurology and the Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine-New York Presbyterian Hospital, New York, NY 10065
| | - Zev Rosenwaks
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Lorenz Studer
- The Biochemistry, Structural Biology, Cell Biology, Developmental Biology and Molecular Biology Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065;
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065;
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
15
|
Peelen DM, Hoogduijn MJ, Hesselink DA, Baan CC. Advanced in vitro Research Models to Study the Role of Endothelial Cells in Solid Organ Transplantation. Front Immunol 2021; 12:607953. [PMID: 33664744 PMCID: PMC7921837 DOI: 10.3389/fimmu.2021.607953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
The endothelium plays a key role in acute and chronic rejection of solid organ transplants. During both processes the endothelium is damaged often with major consequences for organ function. Also, endothelial cells (EC) have antigen-presenting properties and can in this manner initiate and enhance alloreactive immune responses. For decades, knowledge about these roles of EC have been obtained by studying both in vitro and in vivo models. These experimental models poorly imitate the immune response in patients and might explain why the discovery and development of agents that control EC responses is hampered. In recent years, various innovative human 3D in vitro models mimicking in vivo organ structure and function have been developed. These models will extend the knowledge about the diverse roles of EC in allograft rejection and will hopefully lead to discoveries of new targets that are involved in the interactions between the donor organ EC and the recipient's immune system. Moreover, these models can be used to gain a better insight in the mode of action of the currently prescribed immunosuppression and will enhance the development of novel therapeutics aiming to reduce allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Daphne M Peelen
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
16
|
Ibrahim M, Xie B, Richardson MK. The growth of endothelial-like cells in zebrafish embryoid body culture. Exp Cell Res 2020; 392:112032. [PMID: 32353375 DOI: 10.1016/j.yexcr.2020.112032] [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: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 11/25/2022]
Abstract
There is increasing interest in the possibility of culturing organ-like tissues (organoids) in vitro for biomedical applications. The ability to culture organoids would be greatly enhanced by having a functional circulation in vitro. The endothelial cell is the most important cell type in this context. Endothelial cells can be derived from pluripotent embryonic blastocyst cells in aggregates called embryoid bodies. Here, we examine the yield of endothelial-like cells in embryoid bodies (EBs) developed from transgenic zebrafish fli:GFP and kdrl:GFP blastocyst embryos. The isolated blastocyst cells developed into EBs within the first 24 h of culture and contained fli:GFP+ (putative endothelial, hematopoietic and other cell types); or kdrl:GFP+ (endothelial) cells. The addition of endothelial growth supplements to the media and culture on collagen type-I substratum increased the percentages of fli:GFP+ and kdrl:GFP+ cells in culture. We found that EBs developed in hanging-drop cultures possessed a higher percentage of fli:GFP+ (45.0 ± 3.1%) and kdrl:GFP+ cells (8.7 ± 0.7%) than those developed on conventional substrata (34.5 ± 1.4% or 5.2 ± 0.4%, respectively). The transcriptome analysis showed a higher expression of VEGF and TGFβ genes in EB cultures compared to the adherent cultures. When transferred to conventional culture, the percentage of fli:GFP+ or kdrl:GFP+ cells declined significantly over subsequent days in the EBs. The fli:GFP+ cells formed a monolayer around the embryoid bodies, while the kdrl:GFP+ cells formed vascular network-like structures in the embryoid bodies. Differences were observed in the spreading of fli:GFP+ cells, and network formation of kdrl:GFP+ cells on different substrates. The fli:GFP+ cells could be maintained in primary culture and sub-cultures. By contrast, kdrl:GFP+ cells were almost completely absent at 8d of primary culture. Our culture model allows real-time observation of fli:GFP+ and kdrl:GFP+ cells in culture. The results obtained from this study will be important for the development of vascular and endothelial cell culture using embryonic cells.
Collapse
Affiliation(s)
- Muhammad Ibrahim
- Institute of Biology Leiden, Leiden University, The Netherlands; Animal Biotechnology Division, Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Pakistan
| | - Bing Xie
- Institute of Biology Leiden, Leiden University, The Netherlands
| | | |
Collapse
|
17
|
Deng Y, Zhou Z, Lin S, Yu B. METTL1 limits differentiation and functioning of EPCs derived from human-induced pluripotent stem cells through a MAPK/ERK pathway. Biochem Biophys Res Commun 2020; 527:791-798. [PMID: 32430183 DOI: 10.1016/j.bbrc.2020.04.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/22/2020] [Indexed: 01/11/2023]
Abstract
Transplantation of endothelial progenitor cells (EPCs) has high therapeutic potential for ischemia-related ailments like heart attacks and claudication. Due to limited EPC sources, direct reprogramming is a fast-developing way to convert human-induced pluripotent stem cells (hiPSCs) into EPCs fit for transplantation. However, the procedural efficacy was affected by multiple factors, including epigenetic modifications. Recent studies have shown that m7G methylation mediated by Methyltransferase like 1 (METTL1) is required for mouse embryonic stem cells (mESCs) to differentiate normally. Yet, its contributions to EPC differentiation still require elucidation. Here, using immunofluorescence microscopy and Fluorescence-activated Cell Sorting (FACS), we found that the typical EPC markers were significantly increased in METTL1 knockdown (METTL1-KD) hiPSCs-derived EPCs compared to those of control types. In addition, we found that METTL1 knockdown activates the MAPK/ERK signaling pathway during EPCs differentiation from hiPSCs. Furthermore, functional properties of METTL1-KD EPCs were significantly raised above those of control hiPSCs-derived EPCs. Moreover, we proved that METTL1-KD hiPSCs-derived EPCs significantly accelerate vascular smooth muscle cell proliferation and 'phenotype switching' through a co-culture system. To sum up, our results demonstrate that METTL1-KD significantly promotes the differentiation of EPCs along with their in vitro functions, and this effect may be achieved through activation of the MAPK/ERK signaling pathway. This enhances current knowledge of EPC generation from hiPSCs and presents a new therapeutic target of vascular diseases.
Collapse
Affiliation(s)
- Yujie Deng
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhongyang Zhou
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Beixin Yu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
19
|
Palladino A, Mavaro I, Pizzoleo C, De Felice E, Lucini C, de Girolamo P, Netti PA, Attanasio C. Induced Pluripotent Stem Cells as Vasculature Forming Entities. J Clin Med 2019; 8:E1782. [PMID: 31731464 PMCID: PMC6912734 DOI: 10.3390/jcm8111782] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 01/06/2023] Open
Abstract
Tissue engineering (TE) pursues the ambitious goal to heal damaged tissues. One of the most successful TE approaches relies on the use of scaffolds specifically designed and fabricated to promote tissue growth. During regeneration the guidance of biological events may be essential to sustain vasculature neoformation inside the engineered scaffold. In this context, one of the most effective strategies includes the incorporation of vasculature forming cells, namely endothelial cells (EC), into engineered constructs. However, the most common EC sources currently available, intended as primary cells, are affected by several limitations that make them inappropriate to personalized medicine. Human induced Pluripotent Stem Cells (hiPSC), since the time of their discovery, represent an unprecedented opportunity for regenerative medicine applications. Unfortunately, human induced Pluripotent Stem Cells-Endothelial Cells (hiPSC-ECs) still display significant safety issues. In this work, we reviewed the most effective protocols to induce pluripotency, to generate cells displaying the endothelial phenotype and to perform an efficient and safe cell selection. We also provide noteworthy examples of both in vitro and in vivo applications of hiPSC-ECs in order to highlight their ability to form functional blood vessels. In conclusion, we propose hiPSC-ECs as the preferred source of endothelial cells currently available in the field of personalized regenerative medicine.
Collapse
Affiliation(s)
- Antonio Palladino
- CESMA—Centro Servizi Metrologici e Tecnologici Avanzati, University of Naples Federico II, 80146 Naples, Italy
| | - Isabella Mavaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
| | - Carmela Pizzoleo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
| | - Elena De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
| | - Paolo A. Netti
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Chiara Attanasio
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, I-80137 Naples, Italy
- Interdepartmental Center for Research in Biomaterials (CRIB) University of Naples Federico II, I-80125 Naples, Italy
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
20
|
Suhito IR, Kang ES, Kim DS, Baek S, Park SJ, Moon SH, Luo Z, Lee D, Min J, Kim TH. High density gold nanostructure composites for precise electrochemical detection of human embryonic stem cells in cell mixture. Colloids Surf B Biointerfaces 2019; 180:384-392. [PMID: 31082776 DOI: 10.1016/j.colsurfb.2019.04.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/18/2019] [Accepted: 04/29/2019] [Indexed: 01/10/2023]
Abstract
Precise detection of undifferentiated human pluripotent stem cells (hPSCs) and their entire subsequent elimination are incredibly important in preventing teratoma formations after transplantation. Recently, electrochemical sensing platforms have demonstrated immense potential as a new tool to detect remaining hPSCs in label-free and non-destructive manner. Nevertheless, one of the critical huddles of this electrochemical sensing approach is its low sensitivity since even low concentrations of remaining hPSCs were reported to form teratoma once transplanted. To address this issue, in this study, we report an engineering-based approach to improve the sensitivity of electrochemical sensing platform for hPSC detection. By optimizing the density of gold nanostructure and the matrigel concentration to improve both electro-catalytic property and biocompatibility, the sensitivity of the developed platform toward hESCs detection could reach 12,500 cells/chip, which is close to the known critical concentration of hPSCs (˜10,000 cells) that induce teratoma formation in vivo. Remarkably, the electrochemical signals were not detectable from other types of stem cell-derived endothelial cells (CB-EPCs) even at high concentrations of CB-EPCs (40,000 cells/chip), proving the high selectivity of the developed platform toward hPSC detection. Hence, the developed platform could be highly useful to solve the safety issues that are related with clinical application of hPSC-derived cells.
Collapse
Affiliation(s)
- Intan Rosalina Suhito
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Ee-Seul Kang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Da-Seul Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Seungho Baek
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Soon-Jung Park
- Department of Medicine, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Sung-Hwan Moon
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Zhengtang Luo
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Donghyun Lee
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea; Integrative Research Center for Two-Dimensional Functional Materials, Institute of Interdisciplinary Convergence Research, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
21
|
Epac agonist improves barrier function in iPSC-derived endothelial colony forming cells for whole organ tissue engineering. Biomaterials 2019; 200:25-34. [PMID: 30754017 DOI: 10.1016/j.biomaterials.2019.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/06/2019] [Indexed: 02/03/2023]
Abstract
Whole organ engineering paradigms typically involve repopulating acellular organ scaffolds with recipient-compatible cells, to generate a neo-organ that may provide key physiological functions. In the case of whole lung engineering, functionally endothelialized pulmonary vasculature is critical for establishing a fluid-tight barrier at the level of the alveolus, so that oxygen and carbon dioxide can be exchanged in the organ. We have previously developed a protocol to efficiently seed endothelial cells into the microvascular channels of decellularized lung scaffolds, but fully functional endothelial coverage, in terms of barrier function and resistance to thrombosis, was not achieved. In this study, we investigated whether various small molecules could favorably impact endothelial functionality after seeding into decellularized lung scaffolds. We demonstrated that the Epac-selective cAMP analog 8CPT-2Me-cAMP improves endothelial barrier function in repopulated lung scaffolds. When treated with the Epac agonist, barrier function of human umbilical vein endothelial cells (HUVECs) improved, and was maintained for at least three days, whereas the effect of other tested molecules lasted for only 5 h. Treatment with the Epac agonist re-organized actin structure, and appeared to increase the continuity of junction proteins such as VE-cadherin and ZO1. Blockade of actin polymerization abolished the effect of the Epac agonist on barrier function and actin reorganization, confirming a strong actin-mediated effect. Similarly, after treatment with Epac agonist, the barrier function in iPSC-derived endothelial colony forming cells (ECFCs) was increased and the enhanced barrier was maintained for at least 60 h. After culture in lung scaffolds for 5 days, iPSC-ECFCs maintained their phenotype by expressing CD31, eNOS, vWF, and VE-Cadherin. Treatment with the Epac agonist significantly improved the barrier function of iPSC-ECFC-repopulated lung for at least 6 h. Taken together, these findings demonstrated that Epac-selective 8CPT-2Me-cAMP activation enhanced vascular barrier in iPSC-ECFC-engineered lungs, and may be useful to improve endothelial functionality for whole organ tissue engineering.
Collapse
|
22
|
Karagiannis P, Takahashi K, Saito M, Yoshida Y, Okita K, Watanabe A, Inoue H, Yamashita JK, Todani M, Nakagawa M, Osawa M, Yashiro Y, Yamanaka S, Osafune K. Induced Pluripotent Stem Cells and Their Use in Human Models of Disease and Development. Physiol Rev 2019; 99:79-114. [PMID: 30328784 DOI: 10.1152/physrev.00039.2017] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The discovery of somatic cell nuclear transfer proved that somatic cells can carry the same genetic code as the zygote, and that activating parts of this code are sufficient to reprogram the cell to an early developmental state. The discovery of induced pluripotent stem cells (iPSCs) nearly half a century later provided a molecular mechanism for the reprogramming. The initial creation of iPSCs was accomplished by the ectopic expression of four specific genes (OCT4, KLF4, SOX2, and c-Myc; OSKM). iPSCs have since been acquired from a wide range of cell types and a wide range of species, suggesting a universal molecular mechanism. Furthermore, cells have been reprogrammed to iPSCs using a myriad of methods, although OSKM remains the gold standard. The sources for iPSCs are abundant compared with those for other pluripotent stem cells; thus the use of iPSCs to model the development of tissues, organs, and other systems of the body is increasing. iPSCs also, through the reprogramming of patient samples, are being used to model diseases. Moreover, in the 10 years since the first report, human iPSCs are already the basis for new cell therapies and drug discovery that have reached clinical application. In this review, we examine the generation of iPSCs and their application to disease and development.
Collapse
Affiliation(s)
- Peter Karagiannis
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Kazutoshi Takahashi
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Megumu Saito
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Jun K Yamashita
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Masaya Todani
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Masato Nakagawa
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Mitsujiro Osawa
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Yoshimi Yashiro
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| |
Collapse
|
23
|
Bezenah JR, Rioja AY, Juliar B, Friend N, Putnam AJ. Assessing the ability of human endothelial cells derived from induced-pluripotent stem cells to form functional microvasculature in vivo. Biotechnol Bioeng 2019; 116:415-426. [PMID: 30414271 PMCID: PMC6322937 DOI: 10.1002/bit.26860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/26/2022]
Abstract
Forming functional blood vessel networks is a major clinical challenge in the fields of tissue engineering and therapeutic angiogenesis. Cell-based strategies to promote neovascularization have been widely explored, but cell sourcing remains a significant limitation. Induced-pluripotent stem cell-derived endothelial cells (iPSC-ECs) are a promising, potentially autologous, alternative cell source. However, it is unclear whether iPSC-ECs form the same robust microvasculature in vivo documented for other EC sources. In this study, we utilized a well-established in vivo model, in which ECs (iPSC-EC or human umbilical vein endothelial cells [HUVEC]) were coinjected with normal human lung fibroblasts (NHLFs) and a fibrin matrix into the dorsal flank of severe combined immunodeficiency mice to assess their ability to form functional microvasculature. Qualitatively, iPSC-ECs were capable of vessel formation and perfusion and demonstrated similar vessel morphologies to HUVECs. However, quantitatively, iPSC-ECs exhibited a two-fold reduction in vessel density and a three-fold reduction in the number of perfused vessels compared with HUVECs. Further analysis revealed the presence of collagen-IV and α-smooth muscle actin were significantly lower around iPSC-EC/NHLF vasculature than in HUVEC/NHLF implants, suggesting reduced vessel maturity. Collectively, these results demonstrate the need for increased iPSC-EC maturation for clinical translation to be realized.
Collapse
Affiliation(s)
- Jonathan R. Bezenah
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Ana Y. Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Benjamin Juliar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Nicole Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Andrew J. Putnam
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| |
Collapse
|
24
|
Wang K, Lin RZ, Melero-Martin JM. Bioengineering human vascular networks: trends and directions in endothelial and perivascular cell sources. Cell Mol Life Sci 2019; 76:421-439. [PMID: 30315324 PMCID: PMC6349493 DOI: 10.1007/s00018-018-2939-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
Tissue engineering holds great promise in regenerative medicine. However, the field of tissue engineering faces a myriad of difficulties. A major challenge is the necessity to integrate vascular networks into bioengineered constructs to enable physiological functions including adequate oxygenation, nutrient delivery, and removal of waste products. The last two decades have seen remarkable progress in our collective effort to bioengineer human-specific vascular networks. Studies have included both in vitro and in vivo investigations, and multiple methodologies have found varying degrees of success. What most approaches to bioengineer human vascular networks have in common, however, is the synergistic use of both (1) endothelial cells (ECs)-the cells used to line the lumen of the vascular structures and (2) perivascular cells-usually used to support EC function and provide perivascular stability to the networks. Here, we have highlighted trends in the use of various cellular sources over the last two decades of vascular network bioengineering research. To this end, we comprehensively reviewed all life science and biomedical publications available at the MEDLINE database up to 2018. Emphasis was put on selective studies that definitively used human ECs and were specifically related to bioengineering vascular networks. To facilitate this analysis, all papers were stratified by publication year and then analyzed according to their use of EC and perivascular cell types. This study provides an illustrating discussion on how each alternative source of cells has come to be used in the field. Our intention was to reveal trends and to provide new insights into the trajectory of vascular network bioengineering with regard to cellular sources.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
25
|
Boisson-Vidal C, Benslimane-Ahmim Z, Lokajczyk A, Heymann D, Smadja DM. Osteoprotegerin Induces CD34 + Differentiation in Endothelial Progenitor Cells. Front Med (Lausanne) 2018; 5:331. [PMID: 30538990 PMCID: PMC6277572 DOI: 10.3389/fmed.2018.00331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/12/2018] [Indexed: 11/25/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are the main hypothetical cells that could give rise to vessels and in particular one subtype isolated from peripheral or cord bloods: endothelial colony forming cells (ECFCs). These ECFCs are clonogenic precursors committed to endothelial lineage and have robust vasculogenic properties. However, their low number and poor expansion properties when isolated from human adult bloods, currently limit their use as an autologous cell therapy product. We previously reported that osteoprotegerin (OPG), a well-characterized regulator of bone metabolism, contributes to ischemic tissue revascularization, tumor growth in vivo, and potentiates ECFCs proangiogenic properties through the secretion of SDF-1. The current study investigated the role of OPG in ECFCs differentiation and expansion from cord blood CD34+ cells. OPG increased the number of ECFCs after endothelial differentiation of CD34+ cells, enhancing the time of EPCs colonies initial appearance and the growth kinetic of endothelial cell progeny. OPG-exposed ECFCs expressed higher levels of CD34+ compared to control ECFCs. In conclusion, our findings provide novel insights into OPG in regulation of CD34+ progenitor cells. These results give new opportunities for ex vivo expansion of human ECFCs using OPG as a cell culture component for future ECFC product manufacture according to GMP.
Collapse
Affiliation(s)
- Catherine Boisson-Vidal
- Inserm, UMR_S1140, Faculty of Pharmacy, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Zahia Benslimane-Ahmim
- Inserm, UMR_S1140, Faculty of Pharmacy, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anna Lokajczyk
- Inserm, UMR_S1140, Faculty of Pharmacy, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Dominique Heymann
- Inserm, UMR_S1232, CRCINA, Institut de Cancérologie de l'Ouest, Université Nantes-Angers-Le Mans, Nantes, France
| | - David M Smadja
- Inserm, UMR_S1140, Faculty of Pharmacy, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,AP-HP, Hematology Department, European Georges Pompidou Hospital, Paris, France
| |
Collapse
|
26
|
Sweeney M, Foldes G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front Cardiovasc Med 2018; 5:154. [PMID: 30425990 PMCID: PMC6218412 DOI: 10.3389/fcvm.2018.00154] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022] Open
Abstract
The formation of new blood vessels is a crucial step in the development of any new tissue both during embryogenesis and in vitro models as without sufficient perfusion the tissue will be unable to grow beyond the size where nutrition and oxygenation can be managed by diffusion alone. Endothelial cells are the primary building block of blood vessels and are capable of forming tube like structures independently however they are unable to independently form functional vasculature which is capable of conducting blood flow. This requires support from other structures including supporting perivascular cells and the extracellular matrix. The crosstalk between endothelial cells and perivascular cells is vital in regulating vasculogenesis and angiogenesis and the consequences when this is disrupted can be seen in a variety of congenital and acquired disease states. This review details the mechanisms of vasculogenesis in vivo during embryogenesis and compares this to currently employed in vitro techniques. It also highlights clinical consequences of defects in the endothelial cell-pericyte cross-talk and highlights therapies which are being developed to target this pathway. Improving the understanding of the intricacies of endothelial-pericyte signaling will inform pathophysiology of multiple vascular diseases and allow the development of effective in vitro models to guide drug development and assist with approaches in tissue engineering to develop functional vasculature for regenerative medicine applications.
Collapse
Affiliation(s)
- Mark Sweeney
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gabor Foldes
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
27
|
Rodríguez Gutiérrez D, Eid W, Biason-Lauber A. A Human Gonadal Cell Model From Induced Pluripotent Stem Cells. Front Genet 2018; 9:498. [PMID: 30405703 PMCID: PMC6207579 DOI: 10.3389/fgene.2018.00498] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/05/2018] [Indexed: 11/20/2022] Open
Abstract
Sertoli cells are main players in the male gonads development and their study may shed light on 46,XY disorders of sex development (DSD). Mature primary Sertoli cells are incapable of proliferating in prolonged in vitro cultures and the available Sertoli cell models have several limitations since they derive from mouse or human cancer tissues. We differentiated human fibroblasts (HFs)-derived induced pluripotent stem cells into Sertoli-like cells (SLC) and, in order to characterize this new Sertoli cell model, we performed gene expression analyses by NextGeneration Sequencing techniques. This approach revealed that our putative SLC have reduced expression of pluripotency markers and expressed Sertoli cell markers such as SRY-Related HMG-Box 9 (SOX9), vimentin (VIM), and claudin-11 (CLDN-11). More in detail, the transcriptional profile analysis suggested that these cells are in an early stage of Sertoli cells maturation. Harnessing the power of induced pluripotent stem cells, we were able to generate SLC that show genetic and functional similarities to human Sertoli cells (HSerCs). SLC could become an excellent source of patient-specific Sertoli cells that could be of paramount benefit for both basic research and personalized medicine in sex development and reproductive medicine.
Collapse
Affiliation(s)
| | - Wassim Eid
- Section of Medicine, Endocrinology Division, University of Fribourg, Fribourg, Switzerland.,Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Anna Biason-Lauber
- Section of Medicine, Endocrinology Division, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
28
|
Tsifaki M, Kelaini S, Caines R, Yang C, Margariti A. Regenerating the Cardiovascular System Through Cell Reprogramming; Current Approaches and a Look Into the Future. Front Cardiovasc Med 2018; 5:109. [PMID: 30177971 PMCID: PMC6109758 DOI: 10.3389/fcvm.2018.00109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD), despite the advances of the medical field, remains one of the leading causes of mortality worldwide. Discovering novel treatments based on cell therapy or drugs is critical, and induced pluripotent stem cells (iPS Cells) technology has made it possible to design extensive disease-specific in vitro models. Elucidating the differentiation process challenged our previous knowledge of cell plasticity and capabilities and allows the concept of cell reprogramming technology to be established, which has inspired the creation of both in vitro and in vivo techniques. Patient-specific cell lines provide the opportunity of studying their pathophysiology in vitro, which can lead to novel drug development. At the same time, in vivo models have been designed where in situ transdifferentiation of cell populations into cardiomyocytes or endothelial cells (ECs) give hope toward effective cell therapies. Unfortunately, the efficiency as well as the concerns about the safety of all these methods make it exceedingly difficult to pass to the clinical trial phase. It is our opinion that creating an ex vivo model out of patient-specific cells will be one of the most important goals in the future to help surpass all these hindrances. Thus, in this review we aim to present the current state of research in reprogramming toward the cardiovascular system's regeneration, and showcase how the development and study of a multicellular 3D ex vivo model will improve our fighting chances.
Collapse
Affiliation(s)
- Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
29
|
Li L, Miu KK, Gu S, Cheung HH, Chan WY. Comparison of multi-lineage differentiation of hiPSCs reveals novel miRNAs that regulate lineage specification. Sci Rep 2018; 8:9630. [PMID: 29941943 PMCID: PMC6018499 DOI: 10.1038/s41598-018-27719-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/07/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are known to be crucial players in governing the differentiation of human induced pluripotent stem cells (hiPSCs). Despite their utter importance, identifying key lineage specifiers among the myriads of expressed miRNAs remains challenging. We believe that the current practice in mining miRNA specifiers via delineating dynamic fold-changes only is inadequate. Our study, therefore, provides evidence to pronounce "lineage specificity" as another important attribute to qualify for these lineage specifiers. Adopted hiPSCs were differentiated into representative lineages (hepatic, nephric and neuronal) over all three germ layers whilst the depicted miRNA expression changes compiled into an integrated atlas. We demonstrated inter-lineage analysis shall aid in the identification of key miRNAs with lineage-specificity, while these shortlisted candidates were collectively known as "lineage-specific miRNAs". Subsequently, we followed through the fold-changes along differentiation via computational analysis to identify miR-192 and miR-372-3p, respectively, as representative candidate key miRNAs for the hepatic and nephric lineages. Indeed, functional characterization validated that miR-192 and miR-372-3p regulate lineage differentiation via modulation of the expressions of lineage-specific genes. In summary, our presented miRNA atlas is a resourceful ore for the mining of key miRNAs responsible for lineage specification.
Collapse
Affiliation(s)
- Lu Li
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Kai-Kei Miu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
| | - Shen Gu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- M&H Genetics/Baylor Genetics Laboratories, Baylor College of Medicine, Houston, TX, USA
| | - Hoi-Hung Cheung
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| | - Wai-Yee Chan
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| |
Collapse
|
30
|
Bauman E, Granja PL, Barrias CC. Fetal bovine serum-free culture of endothelial progenitor cells-progress and challenges. J Tissue Eng Regen Med 2018; 12:1567-1578. [PMID: 29701896 DOI: 10.1002/term.2678] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 03/22/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Two decades after the first report on endothelial progenitor cells (EPC), their key role in postnatal vasculogenesis and vascular repair is well established. The therapeutic potential of EPC and their growing use in clinical trials calls for the development of more robust, reproducible, and safer methods for the in vitro expansion and maintenance of these cells. Despite many limitations associated with its usage, fetal bovine serum (FBS) is still widely applied as a cell culture supplement. Although different approaches aiming at establishing FBS-free culture have been developed for many cell types, adequate solutions for endothelial cells, and for EPC in particular, are still scarce, possibly due to the multiple challenges that have to be faced when culturing these cells. In this review, we provide a brief overview on the therapeutic relevance of EPC and critically analyse the available literature on FBS-free endothelial cell culture methods, including xeno-free, serum-free, and chemically defined systems.
Collapse
Affiliation(s)
- E Bauman
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal
| | - P L Granja
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - C C Barrias
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
Kurokawa YK, Yin RT, Shang MR, Shirure VS, Moya ML, George SC. Human Induced Pluripotent Stem Cell-Derived Endothelial Cells for Three-Dimensional Microphysiological Systems. Tissue Eng Part C Methods 2018. [PMID: 28622076 DOI: 10.1089/ten.tec.2017.0133] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microphysiological systems (MPS), or "organ-on-a-chip" platforms, aim to recapitulate in vivo physiology using small-scale in vitro tissue models of human physiology. While significant efforts have been made to create vascularized tissues, most reports utilize primary endothelial cells that hinder reproducibility. In this study, we report the use of human induced pluripotent stem cell-derived endothelial cells (iPS-ECs) in developing three-dimensional (3D) microvascular networks. We established a CDH5-mCherry reporter iPS cell line, which expresses the vascular endothelial (VE)-cadherin fused to mCherry. The iPS-ECs demonstrate physiological functions characteristic of primary endothelial cells in a series of in vitro assays, including permeability, response to shear stress, and the expression of endothelial markers (CD31, von Willibrand factor, and endothelial nitric oxide synthase). The iPS-ECs form stable, perfusable microvessels over the course of 14 days when cultured within 3D microfluidic devices. We also demonstrate that inhibition of TGF-β signaling improves vascular network formation by the iPS-ECs. We conclude that iPS-ECs can be a source of endothelial cells in MPS providing opportunities for human disease modeling and improving the reproducibility of 3D vascular networks.
Collapse
Affiliation(s)
- Yosuke K Kurokawa
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Rose T Yin
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Michael R Shang
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Venktesh S Shirure
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Monica L Moya
- 2 Center for Micro and Nano Technology, Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Steven C George
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
- 3 Department of Energy, Environment, and Chemical Engineering, Washington University in St. Louis , St. Louis, Missouri
| |
Collapse
|
32
|
Klein D. iPSCs-based generation of vascular cells: reprogramming approaches and applications. Cell Mol Life Sci 2018; 75:1411-1433. [PMID: 29243171 PMCID: PMC5852192 DOI: 10.1007/s00018-017-2730-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 12/15/2022]
Abstract
Recent advances in the field of induced pluripotent stem cells (iPSCs) research have opened a new avenue for stem cell-based generation of vascular cells. Based on their growth and differentiation potential, human iPSCs constitute a well-characterized, generally unlimited cell source for the mass generation of lineage- and patient-specific vascular cells without any ethical concerns. Human iPSCs-derived vascular cells are perfectly suited for vascular disease modeling studies because patient-derived iPSCs possess the disease-causing mutation, which might be decisive for full expression of the disease phenotype. The application of vascular cells for autologous cell replacement therapy or vascular engineering derived from immune-compatible iPSCs possesses huge clinical potential, but the large-scale production of vascular-specific lineages for regenerative cell therapies depends on well-defined, highly reproducible culture and differentiation conditions. This review will focus on the different strategies to derive vascular cells from human iPSCs and their applications in regenerative therapy, disease modeling and drug discovery approaches.
Collapse
Affiliation(s)
- Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstr. 173, 45122, Essen, Germany.
| |
Collapse
|
33
|
Bezenah JR, Kong YP, Putnam AJ. Evaluating the potential of endothelial cells derived from human induced pluripotent stem cells to form microvascular networks in 3D cultures. Sci Rep 2018; 8:2671. [PMID: 29422650 PMCID: PMC5805762 DOI: 10.1038/s41598-018-20966-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 01/29/2018] [Indexed: 01/11/2023] Open
Abstract
A major translational challenge in the fields of therapeutic angiogenesis and regenerative medicine is the need to create functional microvasculature. The purpose of this study was to assess whether a potentially autologous endothelial cell (EC) source derived from human induced pluripotent stem cells (iPSC-ECs) can form the same robust, stable microvasculature as previously documented for other sources of ECs. We utilized a well-established in vitro assay, in which endothelial cell-coated (iPSC-EC or HUVEC) beads were co-embedded with fibroblasts in a 3D fibrin matrix to assess their ability to form stable microvessels. iPSC-ECs exhibited a five-fold reduction in capillary network formation compared to HUVECs. Increasing matrix density reduced sprouting, although this effect was attenuated by distributing the NHLFs throughout the matrix. Inhibition of both MMP- and plasmin-mediated fibrinolysis was required to completely block sprouting of both HUVECs and iPSC-ECs. Further analysis revealed MMP-9 expression and activity were significantly lower in iPSC-EC/NHLF co-cultures than in HUVEC/NHLF co-cultures at later time points, which may account for the observed deficiencies in angiogenic sprouting of the iPSC-ECs. Collectively, these findings suggest fundamental differences in EC phenotypes must be better understood to enable the promise and potential of iPSC-ECs for clinical translation to be realized.
Collapse
Affiliation(s)
- Jonathan R Bezenah
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Yen P Kong
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Andrew J Putnam
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
34
|
Kwon BJ, Wang X, Kang ML, You J, Lee SJ, Kim WS, Yoon YS, Park JC, Sung HJ. Design of Polymeric Culture Substrates to Promote Proangiogenic Potential of Stem Cells. Macromol Biosci 2017; 18. [PMID: 29285899 DOI: 10.1002/mabi.201700340] [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: 10/09/2017] [Revised: 11/05/2017] [Indexed: 11/11/2022]
Abstract
Stem cells are a promising cell source for regenerative medicine due to their differentiation and self-renewal capacities. In the field of regenerative medicine and tissue engineering, a variety of biomedical technologies have been tested to improve proangiogenic activities of stem cells. However, their therapeutic effect is found to be limited in the clinic because of cell loss, senescence, and insufficient therapeutic activities. To address this type of issue, advanced techniques for biomaterial synthesis and fabrication have been approached to mimic proangiogenic microenvironment and to direct proangiogenic activities. This review highlights the types of polymers and design strategies that have been studied to promote proangiogenic activities of stem cells. In particular, scaffolds, hydrogels, and surface topographies, as well as insight into their underlying mechanisms to improve proangiogenic activities are the focuses. The strategy to promote angiogenic activities of hMSCs by controlling substrate repellency is introduced, and the future direction is proposed.
Collapse
Affiliation(s)
- Byeong-Ju Kwon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.,Cellbiocontrol Laboratory, Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Xintong Wang
- Revenue Analytics, Inc., Atlanta, GA, 30339, USA
| | - Mi-Lan Kang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jin You
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Shin-Jeong Lee
- Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Won Shik Kim
- Department of Otorhinolaryngology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Young-Sup Yoon
- Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.,Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jong-Chul Park
- Cellbiocontrol Laboratory, Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hak-Joon Sung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
35
|
EIF2S3Y suppresses the pluripotency state and promotes the proliferation of mouse embryonic stem cells. Oncotarget 2017; 7:11321-31. [PMID: 26863630 PMCID: PMC4905476 DOI: 10.18632/oncotarget.7187] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/23/2016] [Indexed: 12/15/2022] Open
Abstract
Eukaryotic translation initiation factor 2, subunit 3, and structural gene Y-linked (EIF2S3Y) is essential for spermatogenesis in mouse models. However, its effect on embryonic stem (ES) cells remains unknown. In our observation, differentiated ES cells showed higher levels of EIF2S3Y. To further elucidate its role in ES cells, we utilized ES-derived EIF2S3Y-overexpressing cells and found that EIF2S3Y down-regulated the pluripotency state of ES cells, which might be explained by decreased histone methylation levels because of reduced levels of ten-eleven translocation 1 (TET1). Moreover, EIF2S3Y-overexpressing cells showed an enhanced proliferation rate, which might be due to increased Cyclin A and Cyclin E levels. This study highlighted novel roles of EIF2S3Y in the pluripotency maintenance and proliferation control of ES cells, which would provide an efficient model to study germ cell generation as well as cancer development using ES cells, thus providing valuable target for clinical applications of ES cells.
Collapse
|
36
|
Lin Y, Gil CH, Yoder MC. Differentiation, Evaluation, and Application of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Arterioscler Thromb Vasc Biol 2017; 37:2014-2025. [PMID: 29025705 DOI: 10.1161/atvbaha.117.309962] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.
Collapse
Affiliation(s)
- Yang Lin
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Chang-Hyun Gil
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis.
| |
Collapse
|
37
|
Ibrahim M, Richardson MK. Beyond organoids: In vitro vasculogenesis and angiogenesis using cells from mammals and zebrafish. Reprod Toxicol 2017; 73:292-311. [PMID: 28697965 DOI: 10.1016/j.reprotox.2017.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022]
Abstract
The ability to culture complex organs is currently an important goal in biomedical research. It is possible to grow organoids (3D organ-like structures) in vitro; however, a major limitation of organoids, and other 3D culture systems, is the lack of a vascular network. Protocols developed for establishing in vitro vascular networks typically use human or rodent cells. A major technical challenge is the culture of functional (perfused) networks. In this rapidly advancing field, some microfluidic devices are now getting close to the goal of an artificially perfused vascular network. Another development is the emergence of the zebrafish as a complementary model to mammals. In this review, we discuss the culture of endothelial cells and vascular networks from mammalian cells, and examine the prospects for using zebrafish cells for this objective. We also look into the future and consider how vascular networks in vitro might be successfully perfused using microfluidic technology.
Collapse
Affiliation(s)
- Muhammad Ibrahim
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands; Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Michael K Richardson
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, The Netherlands.
| |
Collapse
|
38
|
Abstract
Because the heart is a poorly regenerative organ, there has been considerable interest in developing novel cell-based approaches to restore lost contractile function after myocardial infarction (MI). While a wide variety of candidate cell types have been tested in animal MI models, the vast majority of clinical trials have used adult stem cell types, usually derived from bone marrow. These studies have generally yielded disappointing results, an outcome that may reflect in part the limited cardiogenic potential of the adult stem cell sources employed. Post-MI heart failure is ultimately a disease of cardiomyocyte deficiency, so better outcomes may be possible with more cardiogenic approaches that may 'remuscularize' the infarct scar with new, electrically-integrated myocardium. In this review, we summarize work in the field to 'program' exogenous or endogenous cells into such a cardiogenic state, as well as efforts to test their capacity to mediate true heart regeneration.
Collapse
Affiliation(s)
- Rocco Romagnuolo
- Toronto General Research Institute, McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada
| | - Michael A Laflamme
- Toronto General Research Institute, McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
39
|
Isolation, characterization and cold storage of cells isolated from diseased explanted livers. Int J Artif Organs 2017; 40:294-306. [PMID: 28574111 DOI: 10.5301/ijao.5000594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2017] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Livers discarded after standard organ retrieval are commonly used as a cell source for hepatocyte transplantation. Due to the scarcity of organ donors, this leads to a shortage of suitable cells for transplantation. Here, the isolation of liver cells from diseased livers removed during liver transplantation is studied and compared to the isolation of cells from liver specimens obtained during partial liver resection. METHODS Hepatocytes from 20 diseased explanted livers (Ex-group) were isolated, cultured and stored at 4°C for up to 48 hours, and compared to hepatocytes isolated from the normal liver tissue of 14 liver lobe resections (Rx-group). The nonparenchymal cell fraction (NPC) was analyzed by flow cytometry to identify potential liver progenitor cells, and OptiPrep™ (Sigma-Aldrich) density gradient centrifugation was used to enrich the progenitor cells for immediate transplantation. RESULTS There were no differences in viability, cell integrity and metabolic activity in cell culture and survival after cold storage when comparing the hepatocytes from the Rx-group and the Ex-group. In some cases, the latter group showed tendencies of increased resistance to isolation and storage procedures. The NPC of the Ex-group livers contained considerably more EpCAM+ and significantly more CD90+ cells than the Rx-group. Progenitor cell enrichment was not sufficient for clinical application. CONCLUSIONS Hepatocytes isolated from diseased explanted livers showed the essential characteristics of being adequate for cell transplantation. Increased numbers of liver progenitor cells can be isolated from diseased explanted livers. These results support the feasibility of using diseased explanted livers as a cell source for liver cell transplantation.
Collapse
|
40
|
Harding A, Cortez-Toledo E, Magner NL, Beegle JR, Coleal-Bergum DP, Hao D, Wang A, Nolta JA, Zhou P. Highly Efficient Differentiation of Endothelial Cells from Pluripotent Stem Cells Requires the MAPK and the PI3K Pathways. Stem Cells 2017; 35:909-919. [PMID: 28248004 DOI: 10.1002/stem.2577] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells are a promising source of endothelial cells (ECs) for the treatment of vascular diseases. We have developed a robust protocol to differentiate human induced pluripotent stem cells (hiPSCs) and embryonic stem cells (hESCs) into ECs with high purities (94%-97% CD31+ and 78%-83% VE-cadherin+ ) in 8 days without cell sorting. Passaging of these cells yielded a nearly pure population of ECs (99% of CD31+ and 96.8% VE-cadherin+ ). These ECs also expressed other endothelial markers vWF, Tie2, NOS3, and exhibited functions of ECs such as uptake of Dil-acetylated low-density lipoprotein and formation of tubes in vitro or vessels in vivo on matrigel. We found that FGF2, VEGF, and BMP4 synergistically induced early vascular progenitors (VPs) from hiPSC-derived mesodermal cells. The MAPK and PI3K pathways are crucial not only for the initial commitment to vascular lineages but also for the differentiation of vascular progenitors to ECs, most likely through regulation of the ETS family transcription factors, ERG and FLI1. We revealed novel roles of the p38 and JNK MAPK pathways on EC differentiation. Furthermore, inhibition of the ERK pathway markedly promoted the differentiation of smooth muscle cells. Finally, we demonstrate that pluripotent stem cell-derived ECs are capable of forming patent blood vessels that were connected to the host vasculature in the ischemic limbs of immune deficient mice. Thus, we demonstrate that ECs can be efficiently derived from hiPSCs and hESCs, and have great potential for vascular therapy as well as for mechanistic studies of EC differentiation. Stem Cells 2017;35:909-919.
Collapse
Affiliation(s)
- Aja Harding
- Department of Internal Medicine, Stem Cell Program.,Department of Biological Sciences, Humboldt State University, Arcata, California, USA
| | | | | | | | | | - Dake Hao
- Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Aijun Wang
- Department of Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Jan A Nolta
- Department of Internal Medicine, Stem Cell Program
| | - Ping Zhou
- Department of Internal Medicine, Stem Cell Program
| |
Collapse
|
41
|
Generation of functional endothelial cells with progenitor-like features from murine induced pluripotent stem cells. Vascul Pharmacol 2016; 86:94-108. [DOI: 10.1016/j.vph.2016.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/20/2016] [Indexed: 11/19/2022]
|
42
|
Significant improvement of direct reprogramming efficacy of fibroblasts into progenitor endothelial cells by ETV2 and hypoxia. Stem Cell Res Ther 2016; 7:104. [PMID: 27488544 PMCID: PMC4973107 DOI: 10.1186/s13287-016-0368-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/18/2016] [Indexed: 12/22/2022] Open
Abstract
Background Endothelial progenitor cell (EPC) transplantation is a promising therapy for ischemic diseases such as ischemic myocardial infarction and hindlimb ischemia. However, limitation of EPC sources remains a major obstacle. Direct reprogramming has become a powerful tool to produce EPCs from fibroblasts. Some recent efforts successfully directly reprogrammed human fibroblasts into functional EPCs; however, the procedure efficacy was low. This study therefore aimed to improve the efficacy of direct reprogramming of human fibroblasts to functional EPCs. Methods Human fibroblasts isolated from foreskin were directly reprogrammed into EPCs by viral ETV2 transduction. Reprogramming efficacy was improved by culturing transduced fibroblasts in hypoxia conditions (5 % oxygen). Phenotype analyses confirmed that single-factor ETV2 transduction successfully reprogrammed dermal fibroblasts into functional EPCs. Results Hypoxia treatment during the reprogramming procedure increased the efficacy of reprogramming from 1.21 ± 0.61 % in normoxia conditions to 7.52 ± 2.31 % in hypoxia conditions. Induced EPCs in hypoxia conditions exhibited functional EPC phenotypes similar to those in normoxia conditions, such as expression of CD31 and VEGFR2, and expressed endothelial gene profiles similar to human umbilical vascular endothelial cells. These cells also formed capillary-like networks in vitro. Conclusion Our study demonstrates a new simple method to increase the reprogramming efficacy of human fibroblasts to EPCs using ETV2 and hypoxia.
Collapse
|
43
|
de l’Hortet AC, Takeishi K, Guzman-Lepe J, Handa K, Matsubara K, Fukumitsu K, Dorko K, Presnell SC, Yagi H, Soto-Gutierrez A. Liver-Regenerative Transplantation: Regrow and Reset. Am J Transplant 2016; 16:1688-96. [PMID: 26699680 PMCID: PMC4874858 DOI: 10.1111/ajt.13678] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/04/2015] [Accepted: 11/26/2015] [Indexed: 01/25/2023]
Abstract
Liver transplantation, either a partial liver from a living or deceased donor or a whole liver from a deceased donor, is the only curative therapy for severe end-stage liver disease. Only one-third of those on the liver transplant waiting list will be transplanted, and the demand for livers is projected to increase 23% in the next 20 years. Consequently, organ availability is an absolute constraint on the number of liver transplants that can be performed. Regenerative therapies aim to enhance liver tissue repair and regeneration by any means available (cell repopulation, tissue engineering, biomaterials, proteins, small molecules, and genes). Recent experimental work suggests that liver repopulation and engineered liver tissue are best suited to the task if an unlimited availability of functional induced pluripotent stem (iPS)-derived liver cells can be achieved. The derivation of iPS cells by reprogramming cell fate has opened up new lines of investigation, for instance, the generation of iPS-derived xenogeneic organs or the possibility of simply inducing the liver to reprogram its own hepatocyte function after injury. We reviewed current knowledge about liver repopulation, generation of engineered livers and reprogramming of liver function. We also discussed the numerous barriers that have to be overcome for clinical implementation.
Collapse
Affiliation(s)
| | - K. Takeishi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - J. Guzman-Lepe
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - K. Handa
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - K. Matsubara
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - K. Fukumitsu
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K. Dorko
- Organovo Holdings Inc., San Diego, CA
| | | | - H. Yagi
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - A. Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA,Thomas E. Starzl Transplantation Institute and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA,Corresponding author: Alejandro Soto-Gutierrez,
| |
Collapse
|
44
|
Quijano LM, Lynch KM, Allan CH, Badylak SF, Ahsan T. Looking Ahead to Engineering Epimorphic Regeneration of a Human Digit or Limb. TISSUE ENGINEERING. PART B, REVIEWS 2016; 22:251-62. [PMID: 26603349 PMCID: PMC4892205 DOI: 10.1089/ten.teb.2015.0401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023]
Abstract
Approximately 2 million people have had limb amputations in the United States due to disease or injury, with more than 185,000 new amputations every year. The ability to promote epimorphic regeneration, or the regrowth of a biologically based digit or limb, would radically change the prognosis for amputees. This ambitious goal includes the regrowth of a large number of tissues that need to be properly assembled and patterned to create a fully functional structure. We have yet to even identify, let alone address, all the obstacles along the extended progression that limit epimorphic regeneration in humans. This review aims to present introductory fundamentals in epimorphic regeneration to facilitate design and conduct of research from a tissue engineering and regenerative medicine perspective. We describe the clinical scenario of human digit healing, featuring published reports of regenerative potential. We then broadly delineate the processes of epimorphic regeneration in nonmammalian systems and describe a few mammalian regeneration models. We give particular focus to the murine digit tip, which allows for comparative studies of regeneration-competent and regeneration-incompetent outcomes in the same animal. Finally, we describe a few forward-thinking opportunities for promoting epimorphic regeneration in humans.
Collapse
Affiliation(s)
- Lina M. Quijano
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Kristen M. Lynch
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Christopher H. Allan
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, Washington
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tabassum Ahsan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| |
Collapse
|
45
|
Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat Rev Cardiol 2016; 13:333-49. [PMID: 27009425 DOI: 10.1038/nrcardio.2016.36] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.
Collapse
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
46
|
Understanding platelet generation from megakaryocytes: implications for in vitro-derived platelets. Blood 2016; 127:1227-33. [PMID: 26787738 DOI: 10.1182/blood-2015-08-607929] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/30/2015] [Indexed: 12/12/2022] Open
Abstract
Platelets are anucleate cytoplasmic discs derived from megakaryocytes that circulate in the blood and have major roles in hemostasis, thrombosis, inflammation, and vascular biology. Platelet transfusions are required to prevent the potentially life-threatening complications of severe thrombocytopenia seen in a variety of medical settings including cancer therapy, trauma, and sepsis. Platelets used in the clinic are currently donor-derived which is associated with concerns over sufficient availability, quality, and complications due to immunologic and/or infectious issues. To overcome our dependence on donor-derived platelets for transfusion, efforts have been made to generate in vitro-based platelets. Work in this area has advanced our understanding of the complex processes that megakaryocytes must undergo to generate platelets both in vivo and in vitro. This knowledge has also defined the challenges that must be overcome to bring in vitro-based platelet manufacturing to a clinical reality. This review will focus on our understanding of committed megakaryocytes and platelet release in vivo and in vitro, and how this knowledge can guide the development of in vitro-derived platelets for clinical application.
Collapse
|
47
|
Avolio E, Caputo M, Madeddu P. Stem cell therapy and tissue engineering for correction of congenital heart disease. Front Cell Dev Biol 2015; 3:39. [PMID: 26176009 PMCID: PMC4485350 DOI: 10.3389/fcell.2015.00039] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/10/2015] [Indexed: 01/08/2023] Open
Abstract
This review article reports on the new field of stem cell therapy and tissue engineering and its potential on the management of congenital heart disease. To date, stem cell therapy has mainly focused on treatment of ischemic heart disease and heart failure, with initial indication of safety and mild-to-moderate efficacy. Preclinical studies and initial clinical trials suggest that the approach could be uniquely suited for the correction of congenital defects of the heart. The basic concept is to create living material made by cellularized grafts that, once implanted into the heart, grows and remodels in parallel with the recipient organ. This would make a substantial improvement in current clinical management, which often requires repeated surgical corrections for failure of implanted grafts. Different types of stem cells have been considered and the identification of specific cardiac stem cells within the heterogeneous population of mesenchymal and stromal cells offers opportunities for de novo cardiomyogenesis. In addition, endothelial cells and vascular progenitors, including cells with pericyte characteristics, may be necessary to generate efficiently perfused grafts. The implementation of current surgical grafts by stem cell engineering could address the unmet clinical needs of patients with congenital heart defects.
Collapse
Affiliation(s)
- Elisa Avolio
- Division of Experimental Cardiovascular Medicine, School of Clinical Sciences, Bristol Heart Institute, University of Bristol Bristol, UK
| | - Massimo Caputo
- Congenital Heart Surgery, School of Clinical Sciences, Bristol Heart Institute, University of Bristol Bristol, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, School of Clinical Sciences, Bristol Heart Institute, University of Bristol Bristol, UK
| |
Collapse
|