1
|
Chung DJ, Wang CH, Liu PJ, Ng SK, Luo CK, Jwo SH, Li CT, Hsu DY, Fan CC, Wei TT. Targeting CREB-binding protein (CBP) abrogates colorectal cancer stemness through epigenetic regulation of C-MYC. Cancer Gene Ther 2024:10.1038/s41417-024-00838-9. [PMID: 39358564 DOI: 10.1038/s41417-024-00838-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Colorectal cancer (CRC) is a common cancer worldwide with an increasing annual incidence. Cancer stem cells (CSCs) play important roles in the occurrence, development, recurrence, and metastasis of CRC. The molecular mechanism regulating the development of colorectal CSCs remains unclear. The discovery of human induced pluripotent stem cells (hiPSCs) through somatic cell reprogramming has revolutionized the fields of stem cell biology and translational medicine. In the present study, we converted hiPSCs into cancer stem-like cells by culture with conditioned medium (CM) from CRC cells. These transformed cells, termed hiPSC-CSCs, displayed cancer stem-like properties, including a spheroid morphology and the expression of both pluripotency and CSC markers. HiPSC-CSCs showed tumorigenic and metastatic abilities in mouse models. The epithelial-mesenchymal transition phenotype was observed in hiPSC-CSCs, which promoted their migration and angiogenesis. Interestingly, upregulation of C-MYC was observed during the differentiation of hiPSC-CSCs. Mechanistically, CREB binding protein (CBP) bound to the C-MYC promoter, while histone deacetylase 1 and 3 (HDAC1/3) dissociated from the promoter, ultimately leading to an increase in histone acetylation and C-MYC transcriptional activation during the differentiation of hiPSC-CSCs. Pharmacological treatment with a CBP inhibitor or abrogation of CBP expression with a CRISPR/Cas9-based strategy reduced the stemness of hiPSC-CSCs. This study demonstrates for the first time that colorectal CSCs can be generated from hiPSCs. The upregulation of C-MYC via histone acetylation plays a crucial role during the conversion process. Inhibition of CBP is a potential strategy for attenuating the stemness of colorectal CSCs.
Collapse
Affiliation(s)
- Dai-Jung Chung
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chun-Hao Wang
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | - Pin-Jung Liu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shang-Kok Ng
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Cong-Kai Luo
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Si-Han Jwo
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chin-Tzu Li
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Dai-Yi Hsu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chia-Chu Fan
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Tzu-Tang Wei
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program in Chemical Biology and Molecular Biophysics (TIGP-CBMB), Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
2
|
Zhao N, Pessell AF, Zhu N, Searson PC. Tissue-Engineered Microvessels: A Review of Current Engineering Strategies and Applications. Adv Healthc Mater 2024; 13:e2303419. [PMID: 38686434 PMCID: PMC11338730 DOI: 10.1002/adhm.202303419] [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: 10/07/2023] [Revised: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Microvessels, including arterioles, capillaries, and venules, play an important role in regulating blood flow, enabling nutrient and waste exchange, and facilitating immune surveillance. Due to their important roles in maintaining normal function in human tissues, a substantial effort has been devoted to developing tissue-engineered models to study endothelium-related biology and pathology. Various engineering strategies have been developed to recapitulate the structural, cellular, and molecular hallmarks of native human microvessels in vitro. In this review, recent progress in engineering approaches, key components, and culture platforms for tissue-engineered human microvessel models is summarized. Then, tissue-specific models, and the major applications of tissue-engineered microvessels in development, disease modeling, drug screening and delivery, and vascularization in tissue engineering, are reviewed. Finally, future research directions for the field are discussed.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ninghao Zhu
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
3
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
4
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
5
|
Liu W, Zhang C, Jiang F, Tan Y, Qin B. From theory to therapy: a bibliometric and visual study of stem cell advancements in age-related macular degeneration. Cytotherapy 2024; 26:616-631. [PMID: 38483361 DOI: 10.1016/j.jcyt.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AIMS Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, offer groundbreaking therapeutic potential for degenerative diseases and cellular repair. Despite their significance, a comprehensive bibliometric analysis in this field, particularly in relation to age-related macular degeneration (AMD), is yet to be conducted. This study aims to map the foundational and emerging areas in stem cell and AMD research through bibliometric analysis. METHODS This study analyzed articles and reviews on stem cells and AMD from 2000 to 2022, sourced from the Web of Science Core Collection. We used VOSviewer and CiteSpace for analysis and visualization of data pertaining to countries, institutions, authors, journals, references and key words. Statistical analyses were conducted using R language and Microsoft Excel 365. RESULTS In total, 539 publications were included, indicating an increase in global literature on stem cells and AMD from 2000 to 2022. The USA was the leading contributor, with 239 papers and the highest H-index, also the USA had the highest average citation rate per article (59.82). Notably, 50% of the top 10 institutions were from the USA, with the University of California system being the most productive. Key authors included Masayo Takahashi, Michiko Mandai, Dennis Clegg, Pete J. Coffey, Boris Stanzel, and Budd A. Tucker. Investigative Ophthalmology & Visual Science published the majority of relevant papers (n = 27). Key words like "clinical trial," "stem cell therapy," "retinal organoid," and "retinal progenitor cells" were predominant. CONCLUSIONS Research on stem cells and AMD has grown significantly, highlighting the need for increased global cooperation. Current research prioritizes the relationship between "ipsc," "induced pluripotent stem cell," "cell culture," and "human embryonic stem cell." As stem cell culture and safety have advanced, focus has shifted to prognosis and complications post-transplantation, signifying the movement of stem cell research from labs to clinical settings.
Collapse
Affiliation(s)
| | | | | | - Yao Tan
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, China; Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha City, China.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China.
| |
Collapse
|
6
|
Fang L, Cai S, McMullen P, Hsu YC, Chen MYQ, Jiang S. Passivating the Background of Living Microbes with a Zwitterionic Peptide for Therapies. Bioconjug Chem 2024; 35:575-581. [PMID: 38456602 DOI: 10.1021/acs.bioconjchem.3c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Living microbial therapies have been proposed as a course of action for a variety of diseases. However, problematic interactions between the host immune system and the microbial organism present significant clinical concerns. Previously, we developed a genetically encoded superhydrophilic zwitterionic peptide, termed EKP, to mimic low-immunogenic zwitterionic materials, which have been used for the chemical modification of biologics such as protein and nucleic acid drugs to increase their in vivo circulation time and reduce their immunogenicity. Herein, we demonstrate the protective effects of the EKP polypeptide genetically cloaking the surface of Saccharomyces cerevisiae as a model microbe in both in vitro and in vivo systems. First, we show that EKP peptide cloaking suppresses the interactions between yeast cells and their specific antibodies, thereby illustrating its cloaking behavior. Then, we examine the in vitro interactions between EKP peptide surface cloaked yeast cells and murine macrophage cells, which exhibit phagocytotic behavior in the presence of foreign microbes. Our results indicate that EKP cloaking suppresses macrophage interactions and thus reduces phagocytosis. Furthermore, EKP cloaked yeast cells demonstrate a prolonged circulation time in mice in vivo.
Collapse
Affiliation(s)
- Liang Fang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Simian Cai
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Patrick McMullen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Yi-Chen Hsu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Michelle Yi Qin Chen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
7
|
Marder M, Remmert C, Perschel JA, Otgonbayar M, von Toerne C, Hauck S, Bushe J, Feuchtinger A, Sheikh B, Moussus M, Meier M. Stem cell-derived vessels-on-chip for cardiovascular disease modeling. Cell Rep 2024; 43:114008. [PMID: 38536819 DOI: 10.1016/j.celrep.2024.114008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
The metabolic syndrome is accompanied by vascular complications. Human in vitro disease models are hence required to better understand vascular dysfunctions and guide clinical therapies. Here, we engineered an open microfluidic vessel-on-chip platform that integrates human pluripotent stem cell-derived endothelial cells (SC-ECs). The open microfluidic design enables seamless integration with state-of-the-art analytical technologies, including single-cell RNA sequencing, proteomics by mass spectrometry, and high-resolution imaging. Beyond previous systems, we report SC-EC maturation by means of barrier formation, arterial toning, and high nitric oxide synthesis levels under gravity-driven flow. Functionally, we corroborate the hallmarks of early-onset atherosclerosis with low sample volumes and cell numbers under flow conditions by determining proteome and secretome changes in SC-ECs stimulated with oxidized low-density lipoprotein and free fatty acids. More broadly, our organ-on-chip platform enables the modeling of patient-specific human endothelial tissue and has the potential to become a general tool for animal-free vascular research.
Collapse
Affiliation(s)
- Maren Marder
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Caroline Remmert
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Julius A Perschel
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | | | | | - Stefanie Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Judith Bushe
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Bilal Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany; Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Michel Moussus
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany; Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
8
|
Mozneb M, Jenkins A, Sances S, Pohlman S, Workman MJ, West D, Ondatje B, El-Ghazawi K, Woodbury A, Garcia VJ, Patel S, Arzt M, Dezem F, Laperle AH, Moser VA, Ho R, Yucer N, Plummer J, Barrett RJ, Svendsen CN, Sharma A. Multi-lineage heart-chip models drug cardiotoxicity and enhances maturation of human stem cell-derived cardiovascular cells. LAB ON A CHIP 2024; 24:869-881. [PMID: 38252454 DOI: 10.1039/d3lc00745f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Cardiovascular toxicity causes adverse drug reactions and may lead to drug removal from the pharmaceutical market. Cancer therapies can induce life-threatening cardiovascular side effects such as arrhythmias, muscle cell death, or vascular dysfunction. New technologies have enabled cardiotoxic compounds to be identified earlier in drug development. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) and vascular endothelial cells (ECs) can screen for drug-induced alterations in cardiovascular cell function and survival. However, most existing hiPSC models for cardiovascular drug toxicity utilize two-dimensional, immature cells grown in static culture. Improved in vitro models to mechanistically interrogate cardiotoxicity would utilize more adult-like, mature hiPSC-derived cells in an integrated system whereby toxic drugs and protective agents can flow between hiPSC-ECs that represent systemic vasculature and hiPSC-CMs that represent heart muscle (myocardium). Such models would be useful for testing the multi-lineage cardiotoxicities of chemotherapeutic drugs such as VEGFR2/PDGFR-inhibiting tyrosine kinase inhibitors (VPTKIs). Here, we develop a multi-lineage, fully-integrated, cardiovascular organ-chip that can enhance hiPSC-EC and hiPSC-CM functional and genetic maturity, model endothelial barrier permeability, and demonstrate long-term functional stability. This microfluidic organ-chip harbors hiPSC-CMs and hiPSC-ECs on separate channels that can be subjected to active fluid flow and rhythmic biomechanical stretch. We demonstrate the utility of this cardiovascular organ-chip as a predictive platform for evaluating multi-lineage VPTKI toxicity. This study may lead to the development of new modalities for the evaluation and prevention of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amelia Jenkins
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Dylan West
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Briana Ondatje
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Kareem El-Ghazawi
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Amanda Woodbury
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Shachi Patel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felipe Dezem
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alex H Laperle
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - V Alexandra Moser
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Nur Yucer
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Jasmine Plummer
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert J Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
9
|
Ma D, Zhang H, Yin L, Xu H, Wu L, Shaji R, Rezai F, Mulla A, Kaur S, Tan S, Kysela B, Wang Y, Chen Z, Zhao C, Gu Y. Human iPSC-derived endothelial cells promote CNS remyelination via BDNF and mTORC1 pathway. Glia 2024; 72:133-155. [PMID: 37675625 DOI: 10.1002/glia.24466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023]
Abstract
Damage of myelin is a component of many diseases in the central nervous system (CNS). The activation and maturation of the quiescent oligodendrocyte progenitor cells (OPCs) are the crucial cellular processes for CNS remyelination, which is influenced by neuroinflammation in the lesion microenvironment. Endothelial cells derived from human induced pluripotent stem cells (hiPSC-ECs) have shown promise in restoring function in various preclinical animal models. Here we ask whether and whether transplantation of hiPSC-ECs could benefit remyelination in a mouse model of CNS demyelination. Our results show that in vitro, hiPSC-ECs increase OPC proliferation, migration and differentiation via secreted soluble factors including brain-derived neurotrophic factor (BDNF). hiPSC-ECs also promote the survival of oligodendrocyte lineage cells in vitro and in vivo. Transplantation of hiPSC-ECs into a toxin-induced demyelination lesion in mouse corpus callosum (CC) leads to increased density of oligodendrocyte lineage cells and level of myelin in demyelinated area, correlated with a decreased neuroinflammation and an increased proportion of pro-regenerative M2 phenotype in microglia/macrophages. The hiPSC-EC-exposed oligodendrocyte lineage cells showed significant increase in the level of phosphorylated S6 ribosomal protein (pS6) both in vitro and in vivo, indicating an involvement of mTORC1 pathway. These results suggest that hiPSC-ECs may benefit myelin protection and regeneration which providing a potential source of cell therapy for a wide range of diseases and injuries associated with myelin damage.
Collapse
Affiliation(s)
- Dan Ma
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Huiyuan Zhang
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Le Yin
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Hao Xu
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Lida Wu
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Rahul Shaji
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Fatema Rezai
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Ayesha Mulla
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Sukhteerath Kaur
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Shengjiang Tan
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Boris Kysela
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Yilong Wang
- Department of Neurology, Tiantan Hospital Capital Medical University, National Center and National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Chinese Institute for Brain Research, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Yuchun Gu
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| |
Collapse
|
10
|
Murphy AR, Allenby MC. In vitro microvascular engineering approaches and strategies for interstitial tissue integration. Acta Biomater 2023; 171:114-130. [PMID: 37717711 DOI: 10.1016/j.actbio.2023.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
The increasing gap between clinical demand for tissue or organ transplants and the availability of donated tissue highlights the emerging opportunities for lab-grown or synthetically engineered tissue. While the field of tissue engineering has existed for nearly half a century, its clinical translation remains unrealised, in part, due to a limited ability to engineer sufficient vascular supply into fabricated tissue, which is necessary to enable nutrient and waste exchange, prevent cellular necrosis, and support tissue proliferation. Techniques to develop anatomically relevant, functional vascular networks in vitro have made significant progress in the last decade, however, the challenge now remains as to how best incorporate these throughout dense parenchymal tissue-like structures to address diffusion-limited development and allow for the fabrication of large-scale vascularised tissue. This review explores advances made in the laboratory engineering of vasculature structures and summarises recent attempts to integrate vascular networks together with sophisticated in vitro avascular tissue and organ-like structures. STATEMENT OF SIGNIFICANCE: The ability to grow full scale, functional tissue and organs in vitro is primarily limited by an inability to adequately diffuse oxygen and nutrients throughout developing cellularised structures, which generally results from the absence of perfusable vessel networks. Techniques to engineering both perfusable vascular networks and avascular miniaturised organ-like structures have recently increased in complexity, sophistication, and physiological relevance. However, integrating these two essential elements into a single functioning vascularised tissue structure represents a significant spatial and temporal engineering challenge which is yet to be surmounted. Here, we explore a range of vessel morphogenic phenomena essential for tissue-vascular co-development, as well as evaluate a range of recent noteworthy approaches for generating vascularised tissue products in vitro.
Collapse
Affiliation(s)
- A R Murphy
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia
| | - M C Allenby
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia; Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
11
|
Xiong Z, An Q, Chen L, Xiang Y, Li L, Zheng Y. Cell or cell derivative-laden hydrogels for myocardial infarction therapy: from the perspective of cell types. J Mater Chem B 2023; 11:9867-9888. [PMID: 37751281 DOI: 10.1039/d3tb01411h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Myocardial infarction (MI) is a global cardiovascular disease with high mortality and morbidity. To treat acute MI, various therapeutic approaches have been developed, including cells, extracellular vesicles, and biomimetic nanoparticles. However, the clinical application of these therapies is limited due to low cell viability, inadequate targetability, and rapid elimination from cardiac sites. Injectable hydrogels, with their three-dimensional porous structure, can maintain the biomechanical stabilization of hearts and the transplantation activity of cells. However, they cannot regenerate cardiomyocytes or repair broken hearts. A better understanding of the collaborative relationship between hydrogel delivery systems and cell or cell-inspired therapy will facilitate advancing innovative therapeutic strategies against MI. Following that, from the perspective of cell types, MI progression and recent studies on using hydrogel to deliver cell or cell-derived preparations for MI treatment are discussed. Finally, current challenges and future prospects of cell or cell derivative-laden hydrogels for MI therapy are proposed.
Collapse
Affiliation(s)
- Ziqing Xiong
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yucheng Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China.
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Sánchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells 2023; 12:2200. [PMID: 37681932 PMCID: PMC10487005 DOI: 10.3390/cells12172200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Disturbances in bone morphogenetic protein (BMP) signalling contribute to onset and development of a number of rare genetic diseases, including Fibrodysplasia ossificans progressiva (FOP), Pulmonary arterial hypertension (PAH), and Hereditary haemorrhagic telangiectasia (HHT). After decades of animal research to build a solid foundation in understanding the underlying molecular mechanisms, the progressive implementation of iPSC-based patient-derived models will improve drug development by addressing drug efficacy, specificity, and toxicity in a complex humanized environment. We will review the current state of literature on iPSC-derived model systems in this field, with special emphasis on the access to patient source material and the complications that may come with it. Given the essential role of BMPs during embryonic development and stem cell differentiation, gain- or loss-of-function mutations in the BMP signalling pathway may compromise iPSC generation, maintenance, and differentiation procedures. This review highlights the need for careful optimization of the protocols used. Finally, we will discuss recent developments towards complex in vitro culture models aiming to resemble specific tissue microenvironments with multi-faceted cellular inputs, such as cell mechanics and ECM together with organoids, organ-on-chip, and microfluidic technologies.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), ISPA-HUCA, Avda. de Roma, s/n, 33011 Oviedo, Spain
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Christian Hiepen
- Department of Engineering and Natural Sciences, Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665 Recklinghausen, Germany
| |
Collapse
|
13
|
Bertucci T, Kakarla S, Winkelman MA, Lane K, Stevens K, Lotz S, Grath A, James D, Temple S, Dai G. Direct differentiation of human pluripotent stem cells into vascular network along with supporting mural cells. APL Bioeng 2023; 7:036107. [PMID: 37564277 PMCID: PMC10411996 DOI: 10.1063/5.0155207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
During embryonic development, endothelial cells (ECs) undergo vasculogenesis to form a primitive plexus and assemble into networks comprised of mural cell-stabilized vessels with molecularly distinct artery and vein signatures. This organized vasculature is established prior to the initiation of blood flow and depends on a sequence of complex signaling events elucidated primarily in animal models, but less studied and understood in humans. Here, we have developed a simple vascular differentiation protocol for human pluripotent stem cells that generates ECs, pericytes, and smooth muscle cells simultaneously. When this protocol is applied in a 3D hydrogel, we demonstrate that it recapitulates the dynamic processes of early human vessel formation, including acquisition of distinct arterial and venous fates, resulting in a vasculogenesis angiogenesis model plexus (VAMP). The VAMP captures the major stages of vasculogenesis, angiogenesis, and vascular network formation and is a simple, rapid, scalable model system for studying early human vascular development in vitro.
Collapse
Affiliation(s)
| | - Shravani Kakarla
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Max A. Winkelman
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Keith Lane
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | | | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | - Alexander Grath
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| | - Daylon James
- Weill Cornell Medicine, New York, New York 10065, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | - Guohao Dai
- Northeastern University, Department of Bioengineering, Boston, Massachusetts 02115, USA
| |
Collapse
|
14
|
Nguyen V, Gao C, Hochman ML, Kravitz J, Chen EH, Friedman HI, Wenceslau CF, Chen D, Wang Y, Nelson JS, Jegga AG, Tan W. Supporting materials: Endothelial cells differentiated from patient dermal fibroblast-derived induced pluripotent stem cells resemble vascular malformations of Port Wine Birthmark. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.02.547408. [PMID: 37662218 PMCID: PMC10473620 DOI: 10.1101/2023.07.02.547408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background Port wine birthmark (PWB) is a congenital vascular malformation resulting from developmentally defective endothelial cells (ECs). Developing clinically relevant disease models for PWB studies is currently an unmet need. Objective Our study aims to generate PWB-derived induced pluripotent stem cells (iPSCs) and iPSC-derived ECs that preserve disease-related phenotypes. Methods PWB iPSCs were generated by reprogramming lesional dermal fibroblasts and differentiated into ECs. RNA-seq was performed to identify differentially expressed genes (DEGs) and enriched pathways. The functional phenotypes of iPSC-derived ECs were characterized by capillary-like structure (CLS) formation in vitro and Geltrex plug-in assay in vivo . Results Human PWB and control iPSC lines were generated through reprogramming of dermal fibroblasts by introducing the "Yamanaka factors" (Oct3/4, Sox2, Klf4, c-Myc) into them; the iPSCs were successfully differentiated into ECs. These iPSCs and their derived ECs were validated by expression of a series of stem cell and EC biomarkers, respectively. PWB iPSC-derived ECs showed impaired CLS in vitro with larger perimeters and thicker branches as compared to control iPSC-derived ECs. In the plug-in assay, perfused human vasculature formed by PWB iPSC- derived ECs showed bigger perimeters and greater densities than those formed by control iPSC- derived ECs in severe combined immune deficient (SCID) mice. The transcriptome analysis showed that dysregulated pathways of stem cell differentiation, Hippo, Wnt, and focal adhesion persisted through differentiation of PWB iPSCs to ECs. Functional enrichment analysis showed that Hippo and Wnt pathway-related PWB DEGs are enriched for vasculature development, tube morphology, endothelium development, and EC differentiation. Further, members of the zinc finger (ZNF) gene family were overrepresented among the DEGs in PWB iPSCs. ZNF DEGs confer significant functions in transcriptional regulation, chromatin remodeling, protein ubiquitination, and retinoic acid receptor signaling. Furthermore, NF-kappa B, TNF, MAPK, and cholesterol metabolism pathways were dysregulated in PWB ECs as readouts of impaired differentiation. Conclusions PWB iPSC-derived ECs render a novel and clinically-relevant disease model by retaining pathological phenotypes. Our data demonstrate multiple pathways, such as Hippo and Wnt, NF-kappa B, TNF, MAPK, and cholesterol metabolism, are dysregulated, which may contribute to the development of differentiation-defective ECs in PWB. Bulleted statements What is already known about this topic?: Port Wine Birthmark (PWB) is a congenital vascular malformation with an incidence rate of 0.1 - 0.3 % per live births.PWB results from developmental defects in the dermal vasculature; PWB endothelial cells (ECs) have differentiational impairments.Pulse dye laser (PDL) is currently the preferred treatment for PWB; unfortunately, the efficacy of PDL treatment of PWB has not improved over the past three decades.What does this study add?: Induced pluripotent stem cells (iPSCs) were generated from PWB skin fibroblasts and differentiated into ECs.PWB ECs recapitulated their pathological phenotypes such as forming enlarged blood vessels in vitro and in vivo.Hippo and Wnt pathways were dysregulated in PWB iPSCs and ECs.Zinc-finger family genes were overrepresented among the differentially expressed genes in PWB iPSCs.Dysregulated NF-kappa B, TNF, MAPK, and cholesterol metabolism pathways were enriched in PWB ECs.What is the translational message?: Targeting Hippo and Wnt pathways and Zinc-finger family genes could restore the physiological differentiation of ECs.Targeting NF-kappa B, TNF, MAPK, and cholesterol metabolism pathways could mitigate the pathological progression of PWB.These mechanisms may lead to the development of paradigm-shifting therapeutic interventions for PWB.
Collapse
Affiliation(s)
- Vi Nguyen
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, USA
| | - Chao Gao
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, USA
| | - Marcelo L Hochman
- The Facial Surgery Center and the Hemangioma & Malformation Treatment Center, Charleston, South Carolina 29425, USA
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425 USA
| | - Jacob Kravitz
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, USA
| | - Elliott H Chen
- Division of Plastic Surgery, School of Medicine, University of South Carolina, Columbia, South Carolina 29203, USA
- Division of Plastic Surgery, Prisma Health Medical Group, Columbia, South Carolina 29203, USA
| | - Harold I Friedman
- Division of Plastic Surgery, School of Medicine, University of South Carolina, Columbia, South Carolina 29203, USA
- Division of Plastic Surgery, Prisma Health Medical Group, Columbia, South Carolina 29203, USA
| | - Camilla F Wenceslau
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina 29208, USA
| | - Dongbao Chen
- Department of Obstetrics and Gynecology, University of California, Irvine, Irvine, California 92617, USA
| | - Yunguan Wang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
- Division of Gastroenterology, Cincinnati Children Hospital Medical Center, Cincinnati, Ohio 45229, USA
- Division of Human Genetics, Cincinnati Children Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - J Stuart Nelson
- Departments of Surgery and Biomedical Engineering, Beckman Laser Institute and Medical Clinic, University of California, Irvine, Irvine, California 92617, USA
| | - Anil G. Jegga
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
- Division of Biomedical Informatics, Cincinnati Children Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Wenbin Tan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina 29208, USA
| |
Collapse
|
15
|
Noh KM, Park SJ, Moon SH, Jung SY. Extracellular matrix cues regulate the differentiation of pluripotent stem cell-derived endothelial cells. Front Cardiovasc Med 2023; 10:1169331. [PMID: 37435057 PMCID: PMC10330705 DOI: 10.3389/fcvm.2023.1169331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/23/2023] [Indexed: 07/13/2023] Open
Abstract
The generation of endothelial cells (ECs) from human pluripotent stem cells (PSCs) has been a promising approach for treating cardiovascular diseases for several years. Human PSCs, particularly induced pluripotent stem cells (iPSCs), are an attractive source of ECs for cell therapy. Although there is a diversity of methods for endothelial cell differentiation using biochemical factors, such as small molecules and cytokines, the efficiency of EC production varies depending on the type and dose of biochemical factors. Moreover, the protocols in which most EC differentiation studies have been performed were in very unphysiological conditions that do not reflect the microenvironment of native tissue. The microenvironment surrounding stem cells exerts variable biochemical and biomechanical stimuli that can affect stem cell differentiation and behavior. The stiffness and components of the extracellular microenvironment are critical inducers of stem cell behavior and fate specification by sensing the extracellular matrix (ECM) cues, adjusting the cytoskeleton tension, and delivering external signals to the nucleus. Differentiation of stem cells into ECs using a cocktail of biochemical factors has been performed for decades. However, the effects of mechanical stimuli on endothelial cell differentiation remain poorly understood. This review provides an overview of the methods used to differentiate ECs from stem cells by chemical and mechanical stimuli. We also propose the possibility of a novel EC differentiation strategy using a synthetic and natural extracellular matrix.
Collapse
Affiliation(s)
- Kyung Mu Noh
- Stem Cell Research Institute, T&R Biofab Co. Ltd., Seongnam-si, Republic of Korea
| | - Soon-Jung Park
- Stem Cell Research Institute, T&R Biofab Co. Ltd., Seongnam-si, Republic of Korea
| | - Sung-Hwan Moon
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong-si, Republic of Korea
| | - Seok Yun Jung
- Stem Cell Research Institute, T&R Biofab Co. Ltd., Seongnam-si, Republic of Korea
| |
Collapse
|
16
|
Ding F, Wu H, Han X, Jiang X, Xiao Y, Tu Y, Yu M, Lei W, Hu S. The miR-148/152 family contributes to angiogenesis of human pluripotent stem cell- derived endothelial cells by inhibiting MEOX2. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:582-593. [PMID: 37200858 PMCID: PMC10185738 DOI: 10.1016/j.omtn.2023.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/19/2023] [Indexed: 05/20/2023]
Abstract
Human pluripotent stem cell-derived endothelial cells (hPSC-ECs) represent a promising source of human ECs urgently needed for the study of cardiovascular disease mechanisms, cell therapy, and drug screening. This study aims to explore the function and regulatory mechanism of the miR-148/152 family consisting of miR-148a, miR-148b, and miR-152 in hPSC-ECs, so as to provide new targets for improving EC function during the above applications. In comparison with the wild-type (WT) group, miR-148/152 family knockout (TKO) significantly reduced the endothelial differentiation efficiency of human embryonic stem cells (hESCs), and impaired the proliferation, migration, and capillary-like tube formatting abilities of their derived ECs (hESC-ECs). Overexpression of miR-152 partially restored the angiogenic capacity of TKO hESC-ECs. Furthermore, the mesenchyme homeobox 2 (MEOX2) was validated as the direct target of miR-148/152 family. MEOX2 knockdown resulted in partial restoration of the angiogenesis ability of TKO hESC-ECs. The Matrigel plug assay further revealed that the in vivo angiogenic capacity of hESC-ECs was impaired by miR-148/152 family knockout, and increased by miR-152 overexpression. Thus, the miR-148/152 family is crucial for maintaining the angiogenesis ability of hPSC-ECs, and might be used as a target to enhance the functional benefit of EC therapy and promote endogenous revascularization.
Collapse
Affiliation(s)
- Fengyue Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Hongchun Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Xinglong Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Xue Jiang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Yang Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Yuanyuan Tu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Corresponding author: Wei Lei, Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Corresponding author: Shijun Hu, Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou 215000, China.
| |
Collapse
|
17
|
Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell 2023; 30:512-529. [PMID: 37084729 PMCID: PMC10201979 DOI: 10.1016/j.stem.2023.03.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/23/2023]
Abstract
Cell-based therapies are being developed for various neurodegenerative diseases that affect the central nervous system (CNS). Concomitantly, the roles of individual cell types in neurodegenerative pathology are being uncovered by genetic and single-cell studies. With a greater understanding of cellular contributions to health and disease and with the arrival of promising approaches to modulate them, effective therapeutic cell products are now emerging. This review examines how the ability to generate diverse CNS cell types from stem cells, along with a deeper understanding of cell-type-specific functions and pathology, is advancing preclinical development of cell products for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
18
|
Arzt M, Pohlman S, Mozneb M, Sharma A. Chemically Defined Production of Tri-Lineage Human iPSC-Derived Cardiac Spheroids. Curr Protoc 2023; 3:e767. [PMID: 37154466 PMCID: PMC11215757 DOI: 10.1002/cpz1.767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cardiac spheroids derived from human induced pluripotent stem cells (hiPSC-cardiac spheroids) represent a powerful three-dimensional (3D) model for examining cardiac physiology and for drug toxicity screening. Recent advances with self-organizing, multicellular cardiac organoids highlight the capability of directed stem cell differentiation approaches to recapitulate the composition of the human heart in vitro. Using hiPSC-derived cardiomyocytes (hiPSC-CMs), hiPSC-derived endothelial cells (hiPSC-ECs), and hiPSC-derived cardiac fibroblasts (hiPSC-CFs) is advantageous for enabling tri-cellular crosstalk within a multilineage system and for generating patient-specific models. Chemically defined medium containing factors needed to simultaneously maintain hiPSC-CMs, hiPSC-ECs, and hiPSC-CFs is used to produce the spheroid system. In this article, we present protocols to illustrate the methods for conducting small-molecule-mediated differentiations of hiPSCs into cardiomyocytes, endothelial cells, and cardiac fibroblasts, as well as to assemble the fully integrated cardiac spheroids. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Maintenance and expansion of hiPSCs Basic Protocol 2: Differentiation of hiPSCs into cardiomyocytes Basic Protocol 3: Differentiation of hiPSCs into vascular endothelial cells Basic Protocol 4: Differentiation of hiPSCs into cardiac fibroblasts Basic Protocol 5: Production of hiPSC-derived cardiac spheroids.
Collapse
Affiliation(s)
- Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- CIRM Bridges to Stem Cell Research Program, California State University, Channel Islands, CA, USA
| | - Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
19
|
Weber J, Weber M, Feile A, Schlensak C, Avci-Adali M. Development of an In Vitro Blood Vessel Model Using Autologous Endothelial Cells Generated from Footprint-Free hiPSCs to Analyze Interactions of the Endothelium with Blood Cell Components and Vascular Implants. Cells 2023; 12:cells12091217. [PMID: 37174617 PMCID: PMC10177426 DOI: 10.3390/cells12091217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Vascular implants, such as stents, are required to treat arterial stenosis or dilatation. The development of innovative stent materials and coatings, as well as novel preclinical testing strategies, is needed to improve the bio- and hemocompatibility of current stents. In this study, a blood vessel-like polydimethylsiloxane (PDMS) model was established to analyze the interaction of an endothelium with vascular implants, as well as blood-derived cells, in vitro. Using footprint-free human induced pluripotent stem cells (hiPSCs) and subsequent differentiation, functional endothelial cells (ECs) expressing specific markers were generated and used to endothelialize an artificial PDMS lumen. The established model was used to demonstrate the interaction of the created endothelium with blood-derived immune cells, which also allowed for real-time imaging. In addition, a stent was inserted into the endothelialized lumen to analyze the surface endothelialization of stents. In the future, this blood vessel-like model could serve as an in vitro platform to test the influence of vascular implants and coatings on endothelialization and to analyze the interaction of the endothelium with blood cell components.
Collapse
Affiliation(s)
- Josefin Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Adrian Feile
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| |
Collapse
|
20
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Pilz RA, Skowronek D, Mellinger L, Bekeschus S, Felbor U, Rath M. Endothelial Differentiation of CCM1 Knockout iPSCs Triggers the Establishment of a Specific Gene Expression Signature. Int J Mol Sci 2023; 24:ijms24043993. [PMID: 36835400 PMCID: PMC9963194 DOI: 10.3390/ijms24043993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Cerebral cavernous malformation (CCM) is a neurovascular disease that can lead to seizures and stroke-like symptoms. The familial form is caused by a heterozygous germline mutation in either the CCM1, CCM2, or CCM3 gene. While the importance of a second-hit mechanism in CCM development is well established, it is still unclear whether it immediately triggers CCM development or whether additional external factors are required. We here used RNA sequencing to study differential gene expression in CCM1 knockout induced pluripotent stem cells (CCM1-/- iPSCs), early mesoderm progenitor cells (eMPCs), and endothelial-like cells (ECs). Notably, CRISPR/Cas9-mediated inactivation of CCM1 led to hardly any gene expression differences in iPSCs and eMPCs. However, after differentiation into ECs, we found the significant deregulation of signaling pathways well known to be involved in CCM pathogenesis. These data suggest that a microenvironment of proangiogenic cytokines and growth factors can trigger the establishment of a characteristic gene expression signature upon CCM1 inactivation. Consequently, CCM1-/- precursor cells may exist that remain silent until entering the endothelial lineage. Collectively, not only downstream consequences of CCM1 ablation but also supporting factors must be addressed in CCM therapy development.
Collapse
Affiliation(s)
- Robin A. Pilz
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, 17475 Greifswald, Germany
| | - Dariush Skowronek
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, 17475 Greifswald, Germany
| | - Lara Mellinger
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), 17489 Greifswald, Germany
| | - Ute Felbor
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, 17475 Greifswald, Germany
| | - Matthias Rath
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, 17475 Greifswald, Germany
- Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany
- Correspondence: ; Tel.: +49-3834-865396
| |
Collapse
|
22
|
Rezvani M, Vallier L, Guillot A. Modeling Nonalcoholic Fatty Liver Disease in the Dish Using Human-Specific Platforms: Strategies and Limitations. Cell Mol Gastroenterol Hepatol 2023; 15:1135-1145. [PMID: 36740045 PMCID: PMC10031472 DOI: 10.1016/j.jcmgh.2023.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease affecting multiple cell types of the human liver. The high prevalence of NAFLD and the lack of approved therapies increase the demand for reliable models for the preclinical discovery of drug targets. In the last decade, multiple proof-of-principle studies have demonstrated human-specific NAFLD modeling in the dish. These systems have included technologies based on human induced pluripotent stem cell derivatives, liver tissue section cultures, intrahepatic cholangiocyte organoids, and liver-on-a-chip. These platforms differ in functional maturity, multicellularity, scalability, and spatial organization. Identifying an appropriate model for a specific NAFLD-related research question is challenging. Therefore, we review different platforms for their strengths and limitations in modeling NAFLD. To define the fidelity of the current human in vitro NAFLD models in depth, we define disease hallmarks within the NAFLD spectrum that range from steatosis to severe fibroinflammatory tissue injury. We discuss how the most common methods are efficacious in modeling genetic contributions and aspects of the early NAFLD-related tissue response. We also highlight the shortcoming of current models to recapitulate the complexity of inter-organ crosstalk and the chronic process of liver fibrosis-to-cirrhosis that usually takes decades in patients. Importantly, we provide methodological overviews and discuss implementation hurdles (eg, reproducibility or costs) to help choose the most appropriate NAFLD model for the individual research focus: hepatocyte injury, ductular reaction, cellular crosstalk, or other applications. In sum, we highlight current strategies and deficiencies to model NAFLD in the dish and propose a framework for the next generation of human-specific investigations.
Collapse
Affiliation(s)
- Milad Rezvani
- Charité Universitätsmedizin Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Berlin, Germany; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Berlin Institute of Health, Center for Regenerative Therapies (BCRT), Berlin, Germany; Berlin Institute of Health, Clinician-Scientist Program, Berlin, Germany
| | - Ludovic Vallier
- Berlin Institute of Health, Center for Regenerative Therapies (BCRT), Berlin, Germany; Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Adrien Guillot
- Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Department of Hepatology & Gastroenterology, Berlin, Germany.
| |
Collapse
|
23
|
McCloskey MC, Zhang VZ, Ahmad SD, Walker S, Romanick SS, Awad HA, McGrath JL. Sourcing cells for in vitro models of human vascular barriers of inflammation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:979768. [PMID: 36483299 PMCID: PMC9724237 DOI: 10.3389/fmedt.2022.979768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 07/20/2023] Open
Abstract
The vascular system plays a critical role in the progression and resolution of inflammation. The contributions of the vascular endothelium to these processes, however, vary with tissue and disease state. Recently, tissue chip models have emerged as promising tools to understand human disease and for the development of personalized medicine approaches. Inclusion of a vascular component within these platforms is critical for properly evaluating most diseases, but many models to date use "generic" endothelial cells, which can preclude the identification of biomedically meaningful pathways and mechanisms. As the knowledge of vascular heterogeneity and immune cell trafficking throughout the body advances, tissue chip models should also advance to incorporate tissue-specific cells where possible. Here, we discuss the known heterogeneity of leukocyte trafficking in vascular beds of some commonly modeled tissues. We comment on the availability of different tissue-specific cell sources for endothelial cells and pericytes, with a focus on stem cell sources for the full realization of personalized medicine. We discuss sources available for the immune cells needed to model inflammatory processes and the findings of tissue chip models that have used the cells to studying transmigration.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samantha S. Romanick
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
24
|
Challenges and opportunities for the next generation of cardiovascular tissue engineering. Nat Methods 2022; 19:1064-1071. [PMID: 36064773 DOI: 10.1038/s41592-022-01591-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 07/07/2022] [Indexed: 12/21/2022]
Abstract
Engineered cardiac tissues derived from human induced pluripotent stem cells offer unique opportunities for patient-specific disease modeling, drug discovery and cardiac repair. Since the first engineered hearts were introduced over two decades ago, human induced pluripotent stem cell-based three-dimensional cardiac organoids and heart-on-a-chip systems have now become mainstays in basic cardiovascular research as valuable platforms for investigating fundamental human pathophysiology and development. However, major obstacles remain to be addressed before the field can truly advance toward commercial and clinical translation. Here we provide a snapshot of the state-of-the-art methods in cardiac tissue engineering, with a focus on in vitro models of the human heart. Looking ahead, we discuss major challenges and opportunities in the field and suggest strategies for enabling broad acceptance of engineered cardiac tissues as models of cardiac pathophysiology and testbeds for the development of therapies.
Collapse
|
25
|
Montero-Calle P, Flandes-Iparraguirre M, Mountris K, S de la Nava A, Laita N, Rosales RM, Iglesias-García O, De-Juan-Pardo EM, Atienza F, Fernández-Santos ME, Peña E, Doblaré M, Gavira JJ, Fernández-Avilés F, Prosper F, Pueyo E, Mazo Vega MM. Fabrication of human myocardium using multidimensional modelling of engineered tissues. Biofabrication 2022; 14. [PMID: 36007502 DOI: 10.1088/1758-5090/ac8cb3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/25/2022] [Indexed: 11/12/2022]
Abstract
Biofabrication of human tissues has seen a meteoric growth triggered by recent technical advancements such as human induced pluripotent stem cells (hiPSCs) and additive manufacturing. However, generation of cardiac tissue is still hampered by lack of addequate mechanical properties and crucially by the often unpredictable post-fabrication evolution of biological components. In this study we employ melt electrowriting (MEW) and hiPSC-derived cardiac cells to generate fibre-reinforced human cardiac minitissues. These are thoroughly characterized in order to build computational models and simulations able to predict their post-fabrication evolution. Our results show that MEW-based human minitissues display advanced maturation 28 post-generation, with a significant increase in the expression of cardiac genes such as MYL2, GJA5, SCN5A and the MYH7/MYH6 and MYL2/MYL7 ratios. Human iPSC-cardiomyocytes are significantly more aligned within the MEW-based 3D tissues, as compared to conventional 2D controls, and also display greater expression of CX43. These are also correlated with a more mature functionality in the form of faster conduction velocity. We used these data to develop simulations capable of accurately reproducing the experimental performance. In-depth gauging of the structural disposition (cellular alignment) and intercellular connectivity (CX43) allowed us to develop an improved computational model able to predict the relationship between cardiac cell alignment and functional performance. This study lays down the path for advancing in the development of in silico tools to predict cardiac biofabricated tissue evolution after generation, and maps the route towards more accurate and biomimetic tissue manufacture.
Collapse
Affiliation(s)
| | | | - Konstantinos Mountris
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018 , SPAIN
| | - Ana S de la Nava
- Hospital General Universitario Gregorio Marañón, 46, Dr. Esquerdo, Madrid, Madrid, 28007, SPAIN
| | - Nicolás Laita
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Ricardo M Rosales
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | | | - Elena M De-Juan-Pardo
- Mechanical Engineering, University of Western Australia Faculty of Engineering Computing and Mathematics, M050, B.Block, 1.36, 35 Stirling Highway, Perth, Perth, Western Australia, 6009, AUSTRALIA
| | - Felipe Atienza
- Hospital General Universitario Gregorio Marañón, 46, Dr. Esquerdo st, Madrid, Madrid, 28007, SPAIN
| | | | - Estefanía Peña
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Manuel Doblaré
- Instituto de Investigación en Ingeniería de Aragón, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Juan J Gavira
- Department of Cardiology, Clínica Universidad de Navarra, Pio XII av, Pamplona, 31008, SPAIN
| | | | - Felipe Prosper
- Hematology, Universidad de Navarra, Pio XII, 36, Pamplona, Navarra, 31008, SPAIN
| | - Esther Pueyo
- Instituto de Investigación en Ingeniería de Aragón, Calle Mariano Esquillor s/n, Zaragoza, 50018, SPAIN
| | | |
Collapse
|
26
|
Canjuga D, Hansen C, Halbrügge F, Hann L, Weiß S, Schlensak C, Wendel HP, Avci-Adali M. Improving hemocompatibility of artificial lungs by click conjugation of glycoengineered endothelial cells onto blood-contacting surfaces. BIOMATERIALS ADVANCES 2022; 137:212824. [PMID: 35929239 DOI: 10.1016/j.bioadv.2022.212824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/01/2022] [Accepted: 04/20/2022] [Indexed: 06/15/2023]
Abstract
Artificial lungs, also known as oxygenators, allow adequate oxygenation of the blood in patients with severe respiratory failure and enable patient survival. However, the insufficient hemocompatibility of the current of artificial lungs hampers their long-term use. Therefore, in this study, a novel strategy was developed to efficiently endothelialize blood-contacting surfaces to improve their hemocompatibility. Hollow fiber membranes (HFMs) were functionalized with dibenzylcyclooctyne (DBCO), and endothelial cells were glycoengineered for covalent conjugation to DBCO by a copper-free click reaction. Metabolic glycoengineering using azidoacetylmannosamine-tetraacylated (Ac4ManNAz) resulted in highly efficient functionalization of endothelial cells with azide (N3) molecules on the cell surface without negative impact on cell viability. After 48 h, significantly improved endothelialization was detected on the HFM surfaces functionalized with DBCO compared to unmodified HFMs. Endothelial cells were responsive to inflammatory stimulus and expressed adhesion-promoting molecules (E-selectin, VCAM-1, and ICAM-1). Furthermore, the hemocompatibility of HFMs was analyzed by dynamic incubation with fresh human blood. DBCO-coated and uncoated HFMs showed a comparable hemocompatibility, but the endothelialization of HFMs significantly reduced the activation of blood coagulation and platelets. Interestingly, the incubation of endothelialized HFMs with human blood further reduced the expression of E-selectin and VCAM-1 in endothelial cells. In this study, a highly efficient, cell-compatible method for endothelialization of artificial lungs was established. This click chemistry-based method can be also applied for the endothelialization of other artificial surfaces for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Denis Canjuga
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Caroline Hansen
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Franziska Halbrügge
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Ludmilla Hann
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sarina Weiß
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Hans-Peter Wendel
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany.
| |
Collapse
|
27
|
O’Connor C, Brady E, Zheng Y, Moore E, Stevens KR. Engineering the multiscale complexity of vascular networks. NATURE REVIEWS. MATERIALS 2022; 7:702-716. [PMID: 35669037 PMCID: PMC9154041 DOI: 10.1038/s41578-022-00447-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 05/14/2023]
Abstract
The survival of vertebrate organisms depends on highly regulated delivery of oxygen and nutrients through vascular networks that pervade nearly all tissues in the body. Dysregulation of these vascular networks is implicated in many common human diseases such as hypertension, coronary artery disease, diabetes and cancer. Therefore, engineers have sought to create vascular networks within engineered tissues for applications such as regenerative therapies, human disease modelling and pharmacological testing. Yet engineering vascular networks has historically remained difficult, owing to both incomplete understanding of vascular structure and technical limitations for vascular fabrication. This Review highlights the materials advances that have enabled transformative progress in vascular engineering by ushering in new tools for both visualizing and building vasculature. New methods such as bioprinting, organoids and microfluidic systems are discussed, which have enabled the fabrication of 3D vascular topologies at a cellular scale with lumen perfusion. These approaches to vascular engineering are categorized into technology-driven and nature-driven approaches. Finally, the remaining knowledge gaps, emerging frontiers and opportunities for this field are highlighted, including the steps required to replicate the multiscale complexity of vascular networks found in nature.
Collapse
Affiliation(s)
- Colleen O’Connor
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
| | - Eileen Brady
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA USA
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA USA
| | - Erika Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL USA
| | - Kelly R. Stevens
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA
- Brotman Baty Institute, Seattle, WA USA
| |
Collapse
|
28
|
Macklin BL, Lin YY, Emmerich K, Wisniewski E, Polster BM, Konstantopoulos K, Mumm JS, Gerecht S. Intrinsic epigenetic control of angiogenesis in induced pluripotent stem cell-derived endothelium regulates vascular regeneration. NPJ Regen Med 2022; 7:28. [PMID: 35551465 PMCID: PMC9098630 DOI: 10.1038/s41536-022-00223-w] [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: 11/21/2021] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Human-induced pluripotent stem cell-derived endothelial cells (iECs) provide opportunities to study vascular development and regeneration, develop cardiovascular therapeutics, and engineer model systems for drug screening. The differentiation and characterization of iECs are well established; however, the mechanisms governing their angiogenic phenotype remain unknown. Here, we aimed to determine the angiogenic phenotype of iECs and the regulatory mechanism controlling their regenerative capacity. In a comparative study with HUVECs, we show that iECs increased expression of vascular endothelial growth factor receptor 2 (VEGFR2) mediates their highly angiogenic phenotype via regulation of glycolysis enzymes, filopodia formation, VEGF mediated migration, and robust sprouting. We find that the elevated expression of VEGFR2 is epigenetically regulated via intrinsic acetylation of histone 3 at lysine 27 by histone acetyltransferase P300. Utilizing a zebrafish xenograft model, we demonstrate that the ability of iECs to promote the regeneration of the amputated fin can be modulated by P300 activity. These findings demonstrate how the innate epigenetic status of iECs regulates their phenotype with implications for their therapeutic potential.
Collapse
Affiliation(s)
- Bria L Macklin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emily Wisniewski
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
29
|
Domingues A, Rossi E, Bujko K, Detriche G, Richez U, Blandinieres A, Kucia M, Ratajczak J, Smadja DM, Ratajczak MZ. Human CD34 + very small embryonic-like stem cells can give rise to endothelial colony-forming cells with a multistep differentiation strategy using UM171 and nicotinamide acid. Leukemia 2022; 36:1440-1443. [PMID: 35169243 PMCID: PMC9061289 DOI: 10.1038/s41375-022-01517-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/28/2022] [Indexed: 01/25/2023]
Affiliation(s)
- Alison Domingues
- Stem Cell Biology Program, University of Louisville, Louisville, KY, 40245, USA
| | - Elisa Rossi
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, F-75006, Paris, France
| | - Kamila Bujko
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grégoire Detriche
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, F-75006, Paris, France
- Vascular Medicine Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Hôpital Européen Georges Pompidou, F-75015, Paris, France
| | - Ulysse Richez
- Vascular Medicine Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Hôpital Européen Georges Pompidou, F-75015, Paris, France
| | - Adeline Blandinieres
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, F-75006, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Hôpital Européen Georges Pompidou, F-75015, Paris, France
| | - Magdalena Kucia
- Stem Cell Biology Program, University of Louisville, Louisville, KY, 40245, USA
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Biology Program, University of Louisville, Louisville, KY, 40245, USA
| | - David M Smadja
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, F-75006, Paris, France.
- Vascular Medicine Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Hôpital Européen Georges Pompidou, F-75015, Paris, France.
| | - Mariusz Z Ratajczak
- Stem Cell Biology Program, University of Louisville, Louisville, KY, 40245, USA.
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
30
|
Gil CH, Chakraborty D, Vieira CP, Prasain N, Calzi SL, Fortmann SD, Hu P, Banno K, Jamal M, Huang C, Sielski MS, Lin Y, Huang X, Dupont MD, Floyd JL, Prasad R, Longhini ALF, McGill TJ, Chung HM, Murphy MP, Kotton DN, Boulton ME, Yoder MC, Grant MB. Specific mesoderm subset derived from human pluripotent stem cells ameliorates microvascular pathology in type 2 diabetic mice. SCIENCE ADVANCES 2022; 8:eabm5559. [PMID: 35245116 PMCID: PMC8896785 DOI: 10.1126/sciadv.abm5559] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) were differentiated into a specific mesoderm subset characterized by KDR+CD56+APLNR+ (KNA+) expression. KNA+ cells had high clonal proliferative potential and specification into endothelial colony-forming cell (ECFCs) phenotype. KNA+ cells differentiated into perfused blood vessels when implanted subcutaneously into the flank of nonobese diabetic/severe combined immunodeficient mice and when injected into the vitreous of type 2 diabetic mice (db/db mice). Transcriptomic analysis showed that differentiation of hiPSCs derived from diabetics into KNA+ cells was sufficient to change baseline differences in gene expression caused by the diabetic status and reprogram diabetic cells to a pattern similar to KNA+ cells derived from nondiabetic hiPSCs. Proteomic array studies performed on retinas of db/db mice injected with either control or diabetic donor-derived KNA+ cells showed correction of aberrant signaling in db/db retinas toward normal healthy retina. These data provide "proof of principle" that KNA+ cells restore perfusion and correct vascular dysfunction in db/db mice.
Collapse
Affiliation(s)
- Chang-Hyun Gil
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dibyendu Chakraborty
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Cristiano P. Vieira
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Nutan Prasain
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Astellas Institute for Regenerative Medicine (AIRM), Westborough, MA 01581, USA
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Seth D. Fortmann
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
- Medical Scientist Training Program (MSTP), School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ping Hu
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Kimihiko Banno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Physiology II, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Mohamed Jamal
- Center for Regenerative Medicine, Pulmonary Center, and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Endodontics, Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 00000, UAE
| | - Chao Huang
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Micheli S. Sielski
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Yang Lin
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Zhongshan-Xuhui Hospital and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 310104, China
| | - Mariana D. Dupont
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Jason L. Floyd
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Ana Leda F. Longhini
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
- Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Trevor J. McGill
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Michael P. Murphy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Pulmonary Center, and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Michael E. Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| |
Collapse
|
31
|
Abstract
Chronic skin wounds are commonly found in older individuals who have impaired circulation due to diabetes or are immobilized due to physical disability. Chronic wounds pose a severe burden to the health-care system and are likely to become increasingly prevalent in aging populations. Various treatment approaches exist to help the healing process, although the healed tissue does not generally recapitulate intact skin but rather forms a scar that has inferior mechanical properties and that lacks appendages such as hair or sweat glands. This article describes new experimental avenues for attempting to improve the regenerative response of skin using biophysical techniques as well as biochemical methods, in some cases by trying to harness the potential of stem cells, either endogenous to the host or provided exogenously, to regenerate the skin. These approaches primarily address the local wound environment and should likely be combined with other modalities to address regional and systemic disease, as well as social determinants of health. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA;
| | - Henry C Hsia
- Department of Surgery, Yale University School of Medicine, and Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Raniga K, Vo NTN, Denning C. Differentiation and Characterization of Human Pluripotent Stem Cell-Derived Cardiac Endothelial Cells for In Vitro Applications. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:339-348. [PMID: 35099750 DOI: 10.1007/978-1-0716-2059-5_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Various protocols have been developed to generate endothelial cells for disease modeling, angiogenesis, vascular regeneration, and drug screening. These protocols often require cell sorting, as most differentiation strategies result in a heterogenous population of endothelial cells (ECs). For any given model system, one important consideration is choosing the appropriate EC subtype, as different EC populations have unique molecular signatures.Herein, we describe a protocol for cardiac EC differentiation and a protocol for endothelial cell characterization. This protocol is aimed at investigating differentiation efficiency by measuring endothelial lineage markers, CD31, VE-Cadherin, and VEGFR2 by flow cytometry. Collectively, these protocols comprise the tools required to generate cardiac ECs efficiently and reproducibly from different hPSC lines without the need for cell sorting. Our protocol adds to the panel of hPSCs for cardiac EC differentiation and addresses reproducibility concerns of hPSC-based experiments. The approaches described are also applicable for complex model generation where multiple cardiovascular cell types are involved and may assist in optimizing differentiations for different cell lineages, including cardiomyocytes, cardiac endothelial cells, and cardiac fibroblasts.
Collapse
Affiliation(s)
- Kavita Raniga
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Nguyen T N Vo
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
33
|
Gifre-Renom L, Daems M, Luttun A, Jones EAV. Organ-Specific Endothelial Cell Differentiation and Impact of Microenvironmental Cues on Endothelial Heterogeneity. Int J Mol Sci 2022; 23:ijms23031477. [PMID: 35163400 PMCID: PMC8836165 DOI: 10.3390/ijms23031477] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
Endothelial cells throughout the body are heterogeneous, and this is tightly linked to the specific functions of organs and tissues. Heterogeneity is already determined from development onwards and ranges from arterial/venous specification to microvascular fate determination in organ-specific differentiation. Acknowledging the different phenotypes of endothelial cells and the implications of this diversity is key for the development of more specialized tissue engineering and vascular repair approaches. However, although novel technologies in transcriptomics and proteomics are facilitating the unraveling of vascular bed-specific endothelial cell signatures, still much research is based on the use of insufficiently specialized endothelial cells. Endothelial cells are not only heterogeneous, but their specialized phenotypes are also dynamic and adapt to changes in their microenvironment. During the last decades, strong collaborations between molecular biology, mechanobiology, and computational disciplines have led to a better understanding of how endothelial cells are modulated by their mechanical and biochemical contexts. Yet, because of the use of insufficiently specialized endothelial cells, there is still a huge lack of knowledge in how tissue-specific biomechanical factors determine organ-specific phenotypes. With this review, we want to put the focus on how organ-specific endothelial cell signatures are determined from development onwards and conditioned by their microenvironments during adulthood. We discuss the latest research performed on endothelial cells, pointing out the important implications of mimicking tissue-specific biomechanical cues in culture.
Collapse
Affiliation(s)
- Laia Gifre-Renom
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven (KU Leuven), BE-3000 Leuven, Belgium; (L.G.-R.); (M.D.); (A.L.)
| | - Margo Daems
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven (KU Leuven), BE-3000 Leuven, Belgium; (L.G.-R.); (M.D.); (A.L.)
| | - Aernout Luttun
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven (KU Leuven), BE-3000 Leuven, Belgium; (L.G.-R.); (M.D.); (A.L.)
| | - Elizabeth A. V. Jones
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven (KU Leuven), BE-3000 Leuven, Belgium; (L.G.-R.); (M.D.); (A.L.)
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| |
Collapse
|
34
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
35
|
Chambers SE, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, Medina RJ. Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl Med 2021; 10 Suppl 2:S54-S61. [PMID: 34724714 PMCID: PMC8560200 DOI: 10.1002/sctm.21-0022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic vascular disease is a major cause of mortality and morbidity worldwide, and regeneration of blood vessels in perfusion-deficient tissues is a worthwhile therapeutic goal. The idea of delivering endothelial stem/progenitor cells to repair damaged vasculature, reperfuse hypoxic tissue, prevent cell death, and consequently diminish tissue inflammation and fibrosis has a strong scientific basis and clinical value. Various labs have proposed endothelial stem/progenitor cell candidates. This has created confusion, as there are profound differences between these cell definitions based on isolation methodology, characterization, and reparative biology. Here, a stricter definition based on stem cell biology principles is proposed. Although preclinical studies have often been promising, results from clinical trials have been highly contradictory and served to highlight multiple challenges associated with disappointing therapeutic benefit. This article reviews recent accomplishments in the field and discusses current difficulties when developing endothelial stem cell therapies. Emerging evidence that disputes the classic view of the bone marrow as the source for these cells and supports the vascular wall as the niche for these tissue-resident endothelial stem cells is considered. In addition, novel markers to identify endothelial stem cells, including CD157, EPCR, and CD31low VEGFR2low IL33+ Sox9+ , are described.
Collapse
Affiliation(s)
- Sarah E.J. Chambers
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Varun Pathak
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Edoardo Pedrini
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Lou Soret
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Nicolas Gendron
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Coralie L. Guerin
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Cytometry Platform, Institut CurieParisFrance
| | - Alan W. Stitt
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - David M. Smadja
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Reinhold J. Medina
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| |
Collapse
|
36
|
Antonyshyn JA, Mazzoli V, McFadden MJ, Gramolini AO, Hofer SOP, Simmons CA, Santerre JP. Mitigating the non-specific uptake of immunomagnetic microparticles enables the extraction of endothelium from human fat. Commun Biol 2021; 4:1205. [PMID: 34671074 PMCID: PMC8528810 DOI: 10.1038/s42003-021-02732-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells are among the fundamental building blocks for vascular tissue engineering. However, a clinically viable source of endothelium has continued to elude the field. Here, we demonstrate the feasibility of sourcing autologous endothelium from human fat – an abundant and uniquely dispensable tissue that can be readily harvested with minimally invasive procedures. We investigate the challenges underlying the overgrowth of human adipose tissue-derived microvascular endothelial cells by stromal cells to facilitate the development of a reliable method for their acquisition. Magnet-assisted cell sorting strategies are established to mitigate the non-specific uptake of immunomagnetic microparticles, enabling the enrichment of endothelial cells to purities that prevent their overgrowth by stromal cells. This work delineates a reliable method for acquiring human adipose tissue-derived microvascular endothelial cells in large quantities with high purities that can be readily applied in future vascular tissue engineering applications. Antonyshyn et al. establish a methodology for acquiring human adipose tissue-derived microvascular endothelial cells that can be readily applied in future vascular tissue engineering applications. The authors developed strategies to mitigate the non-specific uptake of immunomagnetic microparticles to facilitate the immunoselection of endothelial cells by magnet-assisted cell sorting.
Collapse
Affiliation(s)
- Jeremy A Antonyshyn
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Vienna Mazzoli
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Meghan J McFadden
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Stefan O P Hofer
- Division of Plastic, Reconstructive, and Aesthetic Surgery, University of Toronto, Toronto, ON, Canada.,Departments of Surgery and Surgical Oncology, University Health Network, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada.,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - J Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada. .,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada. .,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Quan Y, Shan X, Hu M, Jin P, Ma J, Fan J, Yang J, Zhang H, Fan X, Gong Y, Li M, Wang Y. YAP inhibition promotes endothelial cell differentiation from pluripotent stem cell through EC master transcription factor FLI1. J Mol Cell Cardiol 2021; 163:81-96. [PMID: 34666000 DOI: 10.1016/j.yjmcc.2021.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/14/2021] [Accepted: 10/09/2021] [Indexed: 12/18/2022]
Abstract
Endothelial cells (ECs) derived from pluripotent stem cells (PSCs) provide great resource for vascular disease modeling and cell-based regeneration therapy. However, the molecular mechanisms of EC differentiation are not completely understood. In this study, we checked transcriptional profile by microarray and found Hippo pathway is changed and the activity of YAP decreased during mesoderm-mediated EC differentiation from human embryonic stem cells (hESCs). Knockdown of YAP in hESCs promoted both mesoderm and EC differentiation indicating by mesodermal- or EC-specific marker gene expression increased both in mRNA and protein level. In contrast, overexpression of YAP inhibited mesoderm and EC differentiation. Microarray data showed that several key transcription factors of EC differentiation, such as FLI1, ERG, SOX17 are upregulated. Interestingly, knockdown YAP enhanced the expression of these master transcription factors. Bioinformation analysis revealed that TEAD, a YAP binds transcription factors, might regulate the expression of EC master TFs, including FLI1. Luciferase assay confirmed that YAP binds to TEAD1, which would inhibit FLI1 expression. Finally, FLI1 overexpression rescued the effects of YAP overexpression-mediated inhibition of EC differentiation. In conclusion, we revealed the inhibitory effects of YAP on EC differentiation from PSCs, and YAP inhibition might promote expression of master TFs FLI1 for EC commitment through interacting with TEAD1, which might provide an idea for EC differentiation and vascular regeneration via manipulating YAP signaling.
Collapse
Affiliation(s)
- Yingyi Quan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Xiaoqiong Shan
- The Third People's Hospital of Hangzhou. Hangzhou 310009, Zhejiang, China
| | - Minjie Hu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Peifeng Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Jianshe Ma
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Jiwen Yang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Huan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Ming Li
- Cardiac Regeneration Research Institute, School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Yongyu Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| |
Collapse
|
38
|
Xu X, Liao L, Tian W. Strategies of Prevascularization in Tissue Engineering and Regeneration of Craniofacial Tissues. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:464-475. [PMID: 34191620 DOI: 10.1089/ten.teb.2021.0004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Craniofacial tissue defects caused by trauma, developmental malformation, or surgery are critical issues of high incidence, which are harmful to physical and psychological health. Transplantation of engineered tissues or biomaterials is a potential method to repair defects and regenerate the craniofacial tissues. Revascularization is essential to ensure the survival and regeneration of the grafts. Since microvessels play a critical role in blood circulation and substance exchange, the pre-establishment of the microvascular network in transplants provides a technical basis for the successful regeneration of the tissue defect. In this study, we reviewed the recent development of strategies and applications of prevascularization in tissue engineering and regeneration of craniofacial tissues. We focused on the cellular foundation of the in vitro prevascularized microvascular network, the cell source for prevascularization, and the strategies of prevascularization. Several key strategies, including coculture, microspheres, three-dimensional printing and microfluidics, and microscale technology, were summarized and the feasibility of these technologies in the clinical repair of craniofacial defects was discussed.
Collapse
Affiliation(s)
- Xun Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Joshi S, Lee WH, Chen P, Serpooshan V, Yang H. Editorial: 3D Cell Culture Systems for Cardiovascular Tissue Engineering: In vitro Modelling and in vivo Regenerative Therapies. Front Cardiovasc Med 2021; 8:675676. [PMID: 34222371 PMCID: PMC8247440 DOI: 10.3389/fcvm.2021.675676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Sanika Joshi
- Biomedical Engineering, University of North Texas, Denton, TX, United States.,Texas Academy of Mathematics and Science, University of North Texas, Denton, TX, United States
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Pu Chen
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, United States
| | - Huaxiao Yang
- Biomedical Engineering, University of North Texas, Denton, TX, United States
| |
Collapse
|
40
|
Nguyen J, Lin YY, Gerecht S. The next generation of endothelial differentiation: Tissue-specific ECs. Cell Stem Cell 2021; 28:1188-1204. [PMID: 34081899 DOI: 10.1016/j.stem.2021.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) sense and respond to fluid flow and regulate immune cell trafficking in all organs. Despite sharing the same mesodermal origin, ECs exhibit heterogeneous tissue-specific characteristics. Human pluripotent stem cells (hPSCs) can potentially be harnessed to capture this heterogeneity and further elucidate endothelium behavior to satisfy the need for increased accuracy and breadth of disease models and therapeutics. Here, we review current strategies for hPSC differentiation to blood vascular ECs and their maturation into continuous, fenestrated, and sinusoidal tissues. We then discuss the contribution of hPSC-derived ECs to recent advances in organoid development and organ-on-chip approaches.
Collapse
Affiliation(s)
- Jane Nguyen
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
41
|
Su L, Kong X, Loo SJ, Gao Y, Kovalik JP, Su X, Ma J, Ye L. Diabetic Endothelial Cells Differentiated From Patient iPSCs Show Dysregulated Glycine Homeostasis and Senescence Associated Phenotypes. Front Cell Dev Biol 2021; 9:667252. [PMID: 34136485 PMCID: PMC8201091 DOI: 10.3389/fcell.2021.667252] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Induced pluripotent stem cells derived cells (iPSCs) not only can be used for personalized cell transfer therapy, but also can be used for modeling diseases for drug screening and discovery in vitro. Although prior studies have characterized the function of rodent iPSCs derived endothelial cells (ECs) in diabetes or metabolic syndrome, feature phenotypes are largely unknown in hiPSC-ECs from patients with diabetes. Here, we used hiPSC lines from patients with type 2 diabetes mellitus (T2DM) and differentiated them into ECs (dia-hiPSC-ECs). We found that dia-hiPSC-ECs had disrupted glycine homeostasis, increased senescence, and impaired mitochondrial function and angiogenic potential as compared with healthy hiPSC-ECs. These signature phenotypes will be helpful to establish dia-hiPSC-ECs as models of endothelial dysfunction for understanding molecular mechanisms of disease and for identifying and testing new targets for the treatment of endothelial dysfunction in diabetes.
Collapse
Affiliation(s)
- Liping Su
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Sze Jie Loo
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Yu Gao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jean-Paul Kovalik
- Programme in Cardiovascular & Metabolic Disorders, Duke-National University of Singapore, Singapore, Singapore
| | - Xiaofei Su
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lei Ye
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| |
Collapse
|
42
|
Sustained oxidative stress instigates differentiation of cancer stem cells into tumor endothelial cells: Pentose phosphate pathway, reactive oxygen species and autophagy crosstalk. Biomed Pharmacother 2021; 139:111643. [PMID: 33945913 DOI: 10.1016/j.biopha.2021.111643] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor angiogenesis plays a vital role in tumor growth and metastasis. It is proven that in tumor vasculature, endothelial cells (ECs) originate from a small population of cancer cells introduced as cancer stem cells (CSCs). Autophagy has a vital role in ECs differentiation from CSCs and tumor angiogenesis. High levels of reactive oxygen species (ROS) increased autophagy by inhibition of glucose-6-phosphate dehydrogenase (G6PD) and inactivation of the pentose phosphate pathway (PPP). Previously, we suggested that cancer cells initially increase the glycolysis rate when encountering ROS, then the metabolic balance is changed from glycolysis to PPP, following the continuation of oxidative stress. In this study, we investigate the possible role of persistent oxidative stress in the differentiation of CSCs into tumor ECs by relying on the relationship between the ROS, PPP and autophagy. Because tumor angiogenesis plays an important role in the growth and development of cancer, understanding the mechanisms involved in differentiating ECs from CSCs can help find promising treatments for cancer.
Collapse
|
43
|
Cho S, Lee C, Skylar-Scott MA, Heilshorn SC, Wu JC. Reconstructing the heart using iPSCs: Engineering strategies and applications. J Mol Cell Cardiol 2021; 157:56-65. [PMID: 33895197 DOI: 10.1016/j.yjmcc.2021.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have emerged as a key component of cardiac tissue engineering, enabling studies of cardiovascular disease mechanisms, drug responses, and developmental processes in human 3D tissue models assembled from isogenic cells. Since the very first engineered heart tissues were introduced more than two decades ago, a wide array of iPSC-derived cardiac spheroids, organoids, and heart-on-a-chip models have been developed incorporating the latest available technologies and materials. In this review, we will first outline the fundamental biological building blocks required to form a functional unit of cardiac muscle, including iPSC-derived cells differentiated by soluble factors (e.g., small molecules), extracellular matrix scaffolds, and exogenous biophysical maturation cues. We will then summarize the different fabrication approaches and strategies employed to reconstruct the heart in vitro at varying scales and geometries. Finally, we will discuss how these platforms, with continued improvements in scalability and tissue maturity, can contribute to both basic cardiovascular research and clinical applications in the future.
Collapse
Affiliation(s)
- Sangkyun Cho
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Chelsea Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA
| | - Mark A Skylar-Scott
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Betty Irene Moore Children's Heart Center, Stanford Children's Health, Stanford, CA 94025, USA
| | - Sarah C Heilshorn
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Materials Science and Engineering, Stanford University, Stanford, CA 94025, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94025, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94025, USA.
| |
Collapse
|
44
|
A Novel Hypothesis and Characterization to Isolate Microvascular Endothelial Cells Simultaneously with Adipose-Derived Stem Cells from the Human Adipose-Derived Stromal Vascular Fraction. Tissue Eng Regen Med 2021; 18:429-440. [PMID: 33877617 DOI: 10.1007/s13770-021-00332-5] [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: 12/22/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Angiogenesis and vasculogenesis are essential processes for successful tissue regeneration in tissue engineering and regenerative medicine. The adipose-derived stromal vascular fraction (SVF) is not only a source of adipose stem cells (ASC) but also a suitable source of microvascular endothelial cells because it is a rich capillary network. So, we propose a new hypothesis for isolating adipose-derived human microvascular endothelial cells (HMVEC-A) from the SVF and developed a dual isolation system that isolates two cell types from one tissue. METHOD To isolate HMVEC-A, we analyzed the supernatant discarded when ASC is isolated from the adipose-derived SVF. Based on this analysis, we assumed that the SVF adherent to the bottom of the culture plate was divided into two fractions: the stromal fraction as the ASC-rich fraction, and the vascular fraction (VF) as the endothelial cells-rich fraction floating in the culture supernatant. VF isolation was optimized and the efficiency was compared, and the endothelial cells characteristics of HMVEC-A were confirmed by flow cytometric analysis, immunocytochemistry (ICC), a DiI-acetylated low-density lipoprotein (DiI-Ac-LDL) uptake, and in vitro tube formation assay. RESULTS Consistent with the hypothesis, we found a large population of HMVEC-A in the VF and isolated these HMVEC-A by our isolation method. Additionally, this method had higher yields and shorter doubling times than other endothelial cells isolation methods and showed typical morphological and phenotypic characteristics of endothelial cells. CONCLUSION Cells obtained by the method according to our hypothesis can be applied as a useful source for studies such as tissue-to-tissue networks, angiogenesis and tissue regeneration, patient-specific cell therapy, and organoid chips.
Collapse
|
45
|
Huang J, Feng Q, Wang L, Zhou B. Human Pluripotent Stem Cell-Derived Cardiac Cells: Application in Disease Modeling, Cell Therapy, and Drug Discovery. Front Cell Dev Biol 2021; 9:655161. [PMID: 33869218 PMCID: PMC8049435 DOI: 10.3389/fcell.2021.655161] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac diseases are the leading cause of deaths worldwide; however, to date, there has been limited progress in the development of therapeutic options for these conditions. Animal models have been the most extensively studied methods to recapitulate a wide variety of cardiac diseases, but these models exhibit species-specific differences in physiology, metabolism and genetics, which lead to inaccurate and unpredictable drug safety and efficacy results, resulting in drug attrition. The development of human pluripotent stem cell (hPSC) technology in theory guarantees an unlimited source of human cardiac cells. These hPSC-derived cells are not only well suited for traditional two-dimensional (2-D) monoculture, but also applicable to more complex systems, such as three-dimensional (3-D) organoids, tissue engineering and heart on-a-chip. In this review, we discuss the application of hPSCs in heart disease modeling, cell therapy, and next-generation drug discovery. While the hPSC-related technologies still require optimization, their advances hold promise for revolutionizing cell-based therapies and drug discovery.
Collapse
Affiliation(s)
- Juan Huang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
46
|
Nelson EA, Qiu J, Chavkin NW, Hirschi KK. Directed Differentiation of Hemogenic Endothelial Cells from Human Pluripotent Stem Cells. J Vis Exp 2021:10.3791/62391. [PMID: 33871448 PMCID: PMC8675434 DOI: 10.3791/62391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Blood vessels are ubiquitously distributed within all tissues of the body and perform diverse functions. Thus, derivation of mature vascular endothelial cells, which line blood vessel lumens, from human pluripotent stem cells is crucial for a multitude of tissue engineering and regeneration applications. In vivo, primordial endothelial cells are derived from the mesodermal lineage and are specified toward specific subtypes, including arterial, venous, capillary, hemogenic, and lymphatic. Hemogenic endothelial cells are of particular interest because, during development, they give rise to hematopoietic stem and progenitor cells, which then generate all blood lineages throughout life. Thus, creating a system to generate hemogenic endothelial cells in vitro would provide an opportunity to study endothelial-to-hematopoietic transition, and may lead to ex vivo production of human blood products and reduced reliance on human donors. While several protocols exist for the derivation of progenitor and primordial endothelial cells, generation of well-characterized hemogenic endothelial cells from human stem cells has not been described. Here, a method for the derivation of hemogenic endothelial cells from human embryonic stem cells in approximately 1 week is presented: a differentiation protocol with primitive streak cells formed in response to GSK3β inhibitor (CHIR99021), then mesoderm lineage induction mediated by bFGF, followed by primordial endothelial cell development promoted by BMP4 and VEGF-A, and finally hemogenic endothelial cell specification induced by retinoic acid. This protocol yields a well-defined population of hemogenic endothelial cells that can be used to further understand their molecular regulation and endothelial-to-hematopoietic transition, which has the potential to be applied to downstream therapeutic applications.
Collapse
Affiliation(s)
- Elizabeth A Nelson
- Department of Cell Biology, University of Virginia; Cardiovascular Research Center, University of Virginia
| | - Jingyao Qiu
- Department of Medicine, Yale University School of Medicine; Department of Genetics, Yale University School of Medicine
| | - Nicholas W Chavkin
- Department of Cell Biology, University of Virginia; Cardiovascular Research Center, University of Virginia
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia; Cardiovascular Research Center, University of Virginia; Department of Medicine, Yale University School of Medicine; Department of Genetics, Yale University School of Medicine; Yale Cardiovascular Research Center, Yale University School of Medicine;
| |
Collapse
|
47
|
Abutaleb NO, Truskey GA. Differentiation and characterization of human iPSC-derived vascular endothelial cells under physiological shear stress. STAR Protoc 2021; 2:100394. [PMID: 33796871 PMCID: PMC7995664 DOI: 10.1016/j.xpro.2021.100394] [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] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer a potentially unlimited source to generate endothelial cells (ECs) for numerous applications. Here, we describe a 7-day protocol to differentiate up to 55 million vascular endothelial cells (viECs) from 3.5 million human iPSCs using small molecules to regulate specific transcription factors. We also describe a parallel-plate flow chamber system to study EC behavior under physiological shear stress. For complete details on the use and execution of this protocol, please refer to Atchison et al. (2020). A protocol for differentiating vascular endothelial cells (viECs) from human iPSCs Generation of up to 55 million viECs from 3.5 million iPSCs within 7 days Design and use of parallel-plate flow chamber to study EC behavior under flow viECs express EC markers, upregulate flow-sensitive genes, and align to flow direction
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| |
Collapse
|
48
|
Tu C, Cunningham NJ, Zhang M, Wu JC. Human Induced Pluripotent Stem Cells as a Screening Platform for Drug-Induced Vascular Toxicity. Front Pharmacol 2021; 12:613837. [PMID: 33790786 PMCID: PMC8006367 DOI: 10.3389/fphar.2021.613837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/22/2021] [Indexed: 01/02/2023] Open
Abstract
Evaluation of potential vascular injury is an essential part of the safety study during pharmaceutical development. Vascular liability issues are important causes of drug termination during preclinical investigations. Currently, preclinical assessment of vascular toxicity primarily relies on the use of animal models. However, accumulating evidence indicates a significant discrepancy between animal toxicity and human toxicity, casting doubt on the clinical relevance of animal models for such safety studies. While the causes of this discrepancy are expected to be multifactorial, species differences are likely a key factor. Consequently, a human-based model is a desirable solution to this problem, which has been made possible by the advent of human induced pluripotent stem cells (iPSCs). In particular, recent advances in the field now allow the efficient generation of a variety of vascular cells (e.g., endothelial cells, smooth muscle cells, and pericytes) from iPSCs. Using these cells, different vascular models have been established, ranging from simple 2D cultures to highly sophisticated vascular organoids and microfluidic devices. Toxicity testing using these models can recapitulate key aspects of vascular pathology on molecular (e.g., secretion of proinflammatory cytokines), cellular (e.g., cell apoptosis), and in some cases, tissue (e.g., endothelium barrier dysfunction) levels. These encouraging data provide the rationale for continuing efforts in the exploration, optimization, and validation of the iPSC technology in vascular toxicology.
Collapse
Affiliation(s)
- Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Nathan J Cunningham
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Mao Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Stanford University, Stanford, CA, United States.,Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
49
|
Lam CK, Wu JC. Clinical Trial in a Dish: Using Patient-Derived Induced Pluripotent Stem Cells to Identify Risks of Drug-Induced Cardiotoxicity. Arterioscler Thromb Vasc Biol 2021; 41:1019-1031. [PMID: 33472401 PMCID: PMC11006431 DOI: 10.1161/atvbaha.120.314695] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced cardiotoxicity is a significant clinical issue, with many drugs in the market being labeled with warnings on cardiovascular adverse effects. Treatments are often prematurely halted when cardiotoxicity is observed, which limits their therapeutic potential. Moreover, cardiotoxicity is a major reason for abandonment during drug development, reducing available treatment options for diseases and creating a significant financial burden and disincentive for drug developers. Thus, it is important to minimize the cardiotoxic effects of medications that are in use or in development. To this end, identifying patients at a higher risk of developing cardiovascular adverse effects for the drug of interest may be an effective strategy. The discovery of human induced pluripotent stem cells has enabled researchers to generate relevant cell types that retain a patient's own genome and examine patient-specific disease mechanisms, paving the way for precision medicine. Combined with the rapid development of pharmacogenomic analysis, the ability of induced pluripotent stem cell-derivatives to recapitulate patient-specific drug responses provides a powerful platform to identify subsets of patients who are particularly vulnerable to drug-induced cardiotoxicity. In this review, we will discuss the current use of patient-specific induced pluripotent stem cells in identifying populations who are at risk to drug-induced cardiotoxicity and their potential applications in future precision medicine practice. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
50
|
Kennedy CC, Brown EE, Abutaleb NO, Truskey GA. Development and Application of Endothelial Cells Derived From Pluripotent Stem Cells in Microphysiological Systems Models. Front Cardiovasc Med 2021; 8:625016. [PMID: 33659279 PMCID: PMC7917070 DOI: 10.3389/fcvm.2021.625016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/11/2021] [Indexed: 12/02/2022] Open
Abstract
The vascular endothelium is present in all organs and blood vessels, facilitates the exchange of nutrients and waste throughout different organ systems in the body, and sets the tone for healthy vessel function. Mechanosensitive in nature, the endothelium responds to the magnitude and temporal waveform of shear stress in the vessels. Endothelial dysfunction can lead to atherosclerosis and other diseases. Modeling endothelial function and dysfunction in organ systems in vitro, such as the blood-brain barrier and tissue-engineered blood vessels, requires sourcing endothelial cells (ECs) for these biomedical engineering applications. It can be difficult to source primary, easily renewable ECs that possess the function or dysfunction in question. In contrast, human pluripotent stem cells (hPSCs) can be sourced from donors of interest and renewed almost indefinitely. In this review, we highlight how knowledge of vascular EC development in vivo is used to differentiate induced pluripotent stem cells (iPSC) into ECs. We then describe how iPSC-derived ECs are being used currently in in vitro models of organ function and disease and in vivo applications.
Collapse
Affiliation(s)
- Crystal C. Kennedy
- University Program in Genetics and Genomics, Duke University, Durham, NC, United States
| | - Erin E. Brown
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nadia O. Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|