101
|
Hart DA. Sex Differences in Biological Systems and the Conundrum of Menopause: Potential Commonalities in Post-Menopausal Disease Mechanisms. Int J Mol Sci 2022; 23:4119. [PMID: 35456937 PMCID: PMC9026302 DOI: 10.3390/ijms23084119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Sex-specific differences in biology and physiology likely start at the time of conception and progress and mature during the pre-puberty time frame and then during the transitions accompanying puberty. These sex differences are impacted by both genetics and epigenetic alterations during the maturation process, likely for the purpose of preparing for successful reproduction. For females, later in life (~45-50) they undergo another transition leading to a loss of ovarian hormone production at menopause. The reasons for menopause are not clear, but for a subset of females, menopause is accompanied by an increased risk of a number of diseases or conditions that impact a variety of tissues. Most research has mainly focused on the target cells in each of the affected tissues rather than pursue the alternative option that there may be commonalities in the development of these post-menopausal conditions in addition to influences on specific target cells. This review will address some of the potential commonalities presented by an integration of the literature regarding tissue-specific aspects of these post-menopausal conditions and data presented by space flight/microgravity (a condition not anticipated by evolution) that could implicate a loss of a regulatory function of the microvasculature in the risk attached to the affected tissues. Thus, the loss of the integration of the paracrine relationships between endothelial cells of the microvasculature of the tissues affected in the post-menopausal environment could contribute to the risk for post-menopausal diseases/conditions. The validation of this concept could lead to new approaches for interventions to treat post-menopausal conditions, as well as provide new understanding regarding sex-specific biological regulation.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery and Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 4N1, Canada; ; Tel.: +1-403-220-4571
- Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| |
Collapse
|
102
|
Gómez-Salinero JM, Izzo F, Lin Y, Houghton S, Itkin T, Geng F, Bram Y, Adelson RP, Lu TM, Inghirami G, Xiang JZ, Lis R, Redmond D, Schreiner R, Rabbany SY, Landau DA, Schwartz RE, Rafii S. Specification of fetal liver endothelial progenitors to functional zonated adult sinusoids requires c-Maf induction. Cell Stem Cell 2022; 29:593-609.e7. [PMID: 35364013 PMCID: PMC9290393 DOI: 10.1016/j.stem.2022.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/29/2021] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
The liver vascular network is patterned by sinusoidal and hepatocyte co-zonation. How intra-liver vessels acquire their hierarchical specialized functions is unknown. We study heterogeneity of hepatic vascular cells during mouse development through functional and single-cell RNA-sequencing. The acquisition of sinusoidal endothelial cell identity is initiated during early development and completed postnatally, originating from a pool of undifferentiated vascular progenitors at E12. The peri-natal induction of the transcription factor c-Maf is a critical switch for the sinusoidal identity determination. Endothelium-restricted deletion of c-Maf disrupts liver sinusoidal development, aberrantly expands postnatal liver hematopoiesis, promotes excessive postnatal sinusoidal proliferation, and aggravates liver pro-fibrotic sensitivity to chemical insult. Enforced c-Maf overexpression in generic human endothelial cells switches on a liver sinusoidal transcriptional program that maintains hepatocyte function. c-Maf represents an inducible intra-organotypic and niche-responsive molecular determinant of hepatic sinusoidal cell identity and lays the foundation for the strategies for vasculature-driven liver repair.
Collapse
Affiliation(s)
- Jesus Maria Gómez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Franco Izzo
- Division of Hematology and Medical Oncology, Department of Medicine, and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; New York Genome Center, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Yang Lin
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sean Houghton
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tomer Itkin
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Fuqiang Geng
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert P Adelson
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Tyler M Lu
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Raphael Lis
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sina Y Rabbany
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Dan A Landau
- Division of Hematology and Medical Oncology, Department of Medicine, and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; New York Genome Center, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
103
|
Wang Z, Zhao S, Lin X, Chen G, Kang J, Ma Z, Wang Y, Li Z, Xiao X, He A, Xiang D. Application of Organoids in Carcinogenesis Modeling and Tumor Vaccination. Front Oncol 2022; 12:855996. [PMID: 35371988 PMCID: PMC8968694 DOI: 10.3389/fonc.2022.855996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Organoids well recapitulate organ-specific functions from their tissue of origin and remain fundamental aspects of organogenesis. Organoids are widely applied in biomedical research, drug discovery, and regenerative medicine. There are various cultivated organoid systems induced by adult stem cells and pluripotent stem cells, or directly derived from primary tissues. Researchers have drawn inspiration by combination of organoid technology and tissue engineering to produce organoids with more physiological relevance and suitable for translational medicine. This review describes the value of applying organoids for tumorigenesis modeling and tumor vaccination. We summarize the application of organoids in tumor precision medicine. Extant challenges that need to be conquered to make this technology be more feasible and precise are discussed.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shasha Zhao
- State Key Laboratory of Oncogenes and Related Genes, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaolin Lin
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglong Chen
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Jiawei Kang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | | | - Yiming Wang
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Zhi Li
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Xiuying Xiao
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aina He
- Department of Oncology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, The Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
104
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
105
|
Tsaryk R, Yucel N, Leonard EV, Diaz N, Bondareva O, Odenthal-Schnittler M, Arany Z, Vaquerizas JM, Schnittler H, Siekmann AF. Shear stress switches the association of endothelial enhancers from ETV/ETS to KLF transcription factor binding sites. Sci Rep 2022; 12:4795. [PMID: 35314737 PMCID: PMC8938417 DOI: 10.1038/s41598-022-08645-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) lining blood vessels are exposed to mechanical forces, such as shear stress. These forces control many aspects of EC biology, including vascular tone, cell migration and proliferation. Despite a good understanding of the genes responding to shear stress, our insight into the transcriptional regulation of these genes is much more limited. Here, we set out to study alterations in the chromatin landscape of human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress. To do so, we performed ChIP-Seq for H3K27 acetylation, indicative of active enhancer elements and ATAC-Seq to mark regions of open chromatin in addition to RNA-Seq on HUVEC exposed to 6 h of laminar shear stress. Our results show a correlation of gained and lost enhancers with up and downregulated genes, respectively. DNA motif analysis revealed an over-representation of KLF transcription factor (TF) binding sites in gained enhancers, while lost enhancers contained more ETV/ETS motifs. We validated a subset of flow responsive enhancers using luciferase-based reporter constructs and CRISPR-Cas9 mediated genome editing. Lastly, we characterized the shear stress response in ECs of zebrafish embryos using RNA-Seq. Our results lay the groundwork for the exploration of shear stress responsive elements in controlling EC biology.
Collapse
Affiliation(s)
- Roman Tsaryk
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nora Yucel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Elvin V Leonard
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Noelia Diaz
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Olga Bondareva
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103, Leipzig, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Juan M Vaquerizas
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Hans Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany.
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany.
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
106
|
Malhi NK, Luo Y, Tang X, Sriram K, Calandrelli R, Zhong S, Chen ZB. Isolation and Profiling of Human Primary Mesenteric Arterial Endothelial Cells at the Transcriptome Level. J Vis Exp 2022:10.3791/63307. [PMID: 35343966 PMCID: PMC9180814 DOI: 10.3791/63307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023] Open
Abstract
Endothelial cells (ECs) are crucial for vascular and whole-body function through their dynamic response to environmental cues. Elucidating the transcriptome and epigenome of ECs is paramount to understanding their roles in development, health, and disease, but is limited in the availability of isolated primary cells. Recent technologies have enabled the high-throughput profiling of EC transcriptome and epigenome, leading to the identification of previously unknown EC cell subpopulations and developmental trajectories. While EC cultures are a useful tool in the exploration of EC function and dysfunction, the culture conditions and multiple passages can introduce external variables that alter the properties of native EC, including morphology, epigenetic state, and gene expression program. To overcome this limitation, the present paper demonstrates a method of isolating human primary ECs from donor mesenteric arteries aiming to capture their native state. ECs in the intimal layer are dissociated mechanically and biochemically with the use of particular enzymes. The resultant cells can be directly used for bulk RNA or single-cell RNA-sequencing or plated for culture. In addition, a workflow is described for the preparation of human arterial tissue for spatial transcriptomics, specifically for a commercially available platform, although this method is also suitable for other spatial transcriptome profiling techniques. This methodology can be applied to different vessels collected from a variety of donors in health or disease states to gain insights into EC transcriptional and epigenetic regulation, a pivotal aspect of endothelial cell biology.
Collapse
Affiliation(s)
| | - Yingjun Luo
- Department of Diabetes Complications and Metabolism, City of Hope
| | - Xiaofang Tang
- Department of Diabetes Complications and Metabolism, City of Hope
| | - Kiran Sriram
- Department of Diabetes Complications and Metabolism, City of Hope; Irell and Manella Graduate School of Biological Sciences, City of Hope
| | | | - Sheng Zhong
- Department of Bioengineering, University of California San Diego
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, City of Hope; Irell and Manella Graduate School of Biological Sciences, City of Hope;
| |
Collapse
|
107
|
Luo L, Ma Y, Zheng Y, Su J, Huang G. Application Progress of Organoids in Colorectal Cancer. Front Cell Dev Biol 2022; 10:815067. [PMID: 35273961 PMCID: PMC8902504 DOI: 10.3389/fcell.2022.815067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Currently, colorectal cancer is still the third leading cause of cancer-related mortality, and the incidence is rising. It is a long time since the researchers used cancer cell lines and animals as the study subject. However, these models possess various limitations to reflect the cancer progression in the human body. Organoids have more clinical significance than cell lines, and they also bridge the gap between animal models and humans. Patient-derived organoids are three-dimensional cultures that simulate the tumor characteristics in vivo and recapitulate tumor cell heterogeneity. Therefore, the emergence of colorectal cancer organoids provides an unprecedented opportunity for colorectal cancer research. It retains the molecular and cellular composition of the original tumor and has a high degree of homology and complexity with patient tissues. Patient-derived colorectal cancer organoids, as personalized tumor organoids, can more accurately simulate colorectal cancer patients’ occurrence, development, metastasis, and predict drug response in colorectal cancer patients. Colorectal cancer organoids show great potential for application, especially preclinical drug screening and prediction of patient response to selected treatment options. Here, we reviewed the application of colorectal cancer organoids in disease model construction, basic biological research, organoid biobank construction, drug screening and personalized medicine, drug development, drug toxicity and safety, and regenerative medicine. In addition, we also displayed the current limitations and challenges of organoids and discussed the future development direction of organoids in combination with other technologies. Finally, we summarized and analyzed the current clinical trial research of organoids, especially the clinical trials of colorectal cancer organoids. We hoped to lay a solid foundation for organoids used in colorectal cancer research.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yucui Ma
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Yilin Zheng
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| | - Jiating Su
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| |
Collapse
|
108
|
Taelman J, Diaz M, Guiu J. Human Intestinal Organoids: Promise and Challenge. Front Cell Dev Biol 2022; 10:854740. [PMID: 35359445 PMCID: PMC8962662 DOI: 10.3389/fcell.2022.854740] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
The study of human intestinal biology in healthy and diseased conditions has always been challenging. Primary obstacles have included limited tissue accessibility, inadequate in vitro maintenance and ethical constrains. The development of three-dimensional organoid cultures has transformed this entirely. Intestinal organoids are self-organized three-dimensional structures that partially recapitulate the identity, cell heterogeneity and cell behaviour of the original tissue in vitro. This includes the capacity of stem cells to self-renew, as well as to differentiate towards major intestinal lineages. Therefore, over the past decade, the use of human organoid cultures has been instrumental to model human intestinal development, homeostasis, disease, and regeneration. Intestinal organoids can be derived from pluripotent stem cells (PSC) or from adult somatic intestinal stem cells (ISC). Both types of organoid sources harbour their respective strengths and weaknesses. In this mini review, we describe the applications of human intestinal organoids, discussing the differences, advantages, and disadvantages of PSC-derived and ISC-derived organoids.
Collapse
Affiliation(s)
- Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Mònica Diaz
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
- *Correspondence: Jordi Guiu,
| |
Collapse
|
109
|
Generation of αGal-enhanced bifunctional tumor vaccine. Acta Pharm Sin B 2022; 12:3177-3186. [PMID: 35865091 PMCID: PMC9293690 DOI: 10.1016/j.apsb.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with poor prognosis and high mortality. In this study, we demonstrated a novel vaccine targeting HCC and tumor neovascular endothelial cells by fusing recombinant MHCC97H cells expressing porcine α-1,3-galactose epitopes (αGal) and endorphin extracellular domains (END) with dendritic cells (DCs) from healthy volunteers. END+/Gal+-MHCC97H/DC fusion cells induced cytotoxic T lymphocytes (CTLs) and secretion of interferon-gamma (IFN-γ). CTLs targeted cells expressing αGal and END and tumor angiogenesis. The fused cell vaccine can effectively inhibit tumor growth and prolong the survival time of human hepatoma mice, indicating the high clinical potential of this new cell based vaccine.
Collapse
|
110
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
111
|
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: 36] [Impact Index Per Article: 12.0] [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
|
112
|
Li W, Liu JB, Hou LK, Yu F, Zhang J, Wu W, Tang XM, Sun F, Lu HM, Deng J, Bai J, Li J, Wu CY, Lin QL, Lv ZW, Wang GR, Jiang GX, Ma YS, Fu D. Liquid biopsy in lung cancer: significance in diagnostics, prediction, and treatment monitoring. Mol Cancer 2022; 21:25. [PMID: 35057806 PMCID: PMC8772097 DOI: 10.1186/s12943-022-01505-z] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Primary lung cancer is one of the most common malignant tumors in China. Approximately 60% of lung cancer patients have distant metastasis at the initial diagnosis, so it is necessary to find new tumor markers for early diagnosis and individualized treatment. Tumor markers contribute to the early diagnosis of lung cancer and play important roles in early detection and treatment, as well as in precision medicine, efficacy monitoring, and prognosis prediction. The pathological diagnosis of lung cancer in small biopsy specimens determines whether there are tumor cells in the biopsy and tumor type. Because biopsy is traumatic and the compliance of patients with multiple biopsies is poor, liquid biopsy has become a hot research direction. Liquid biopsies are advantageous because they are nontraumatic, easy to obtain, reflect the overall state of the tumor, and allow for real-time monitoring. At present, liquid biopsies mainly include circulating tumor cells, circulating tumor DNA, exosomes, microRNA, circulating RNA, tumor platelets, and tumor endothelial cells. This review introduces the research progress and clinical application prospect of liquid biopsy technology for lung cancer.
Collapse
|
113
|
Itkin T, Rafii S. Cardiovascular diseases disrupt the bone-marrow niche. Nature 2022; 601:515-517. [PMID: 34949859 DOI: 10.1038/d41586-021-03550-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
114
|
Ryan AR, Cleaver O. Plumbing our organs: Lessons from vascular development to instruct lab generated tissues. Curr Top Dev Biol 2022; 148:165-194. [DOI: 10.1016/bs.ctdb.2022.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
115
|
Narayanan S, Vicent S, Ponz-Sarvisé M. PDAC as an Immune Evasive Disease: Can 3D Model Systems Aid to Tackle This Clinical Problem? Front Cell Dev Biol 2021; 9:787249. [PMID: 34957115 PMCID: PMC8703167 DOI: 10.3389/fcell.2021.787249] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with a high mortality rate. The presence of a dense desmoplastic stroma rich in fibroblasts, extracellular matrix, and immune cells plays a critical role in disease progression, therapy response and is a distinguishing feature of PDAC. PDAC is currently treated with a combination of surgery, chemotherapy and radiation therapy in selected cases which results in long-term survival only in a small percentage of patients. Cancer therapies that incorporate immunotherapy-based techniques have become increasingly common in recent years. While such a strategy has been shown to be effective for immunogenic, “hot” tumors like melanoma and lung cancer, thus far PDAC patients display poor responses to this therapeutic approach. Various factors, such as low tumor mutational burden, increased infiltration of immunosuppressive cells, like MDSCs and Treg cells promote tolerance and immune deviation, further aggravating adaptive immunity in PDAC. In this review we will elaborate on the ability of PDAC tumors to evade immune detection. We will also discuss various 3D model system that can be used as a platform in preclinical research to investigate rational combinations of immunotherapy with chemotherapy or targeted therapy, to prime the immune microenvironment to enhance antitumor activity.
Collapse
Affiliation(s)
- Shruthi Narayanan
- Clinica Universidad de Navarra, Medical Oncology Department, Pamplona, Spain
- Program in Solid Tumors, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Silve Vicent
- Program in Solid Tumors, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
- *Correspondence: Silve Vicent, ; Mariano Ponz-Sarvisé,
| | - Mariano Ponz-Sarvisé
- Clinica Universidad de Navarra, Medical Oncology Department, Pamplona, Spain
- Program in Solid Tumors, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
- *Correspondence: Silve Vicent, ; Mariano Ponz-Sarvisé,
| |
Collapse
|
116
|
Developmental angiocrine diversification of endothelial cells for organotypic regeneration. Dev Cell 2021; 56:3042-3051. [PMID: 34813766 DOI: 10.1016/j.devcel.2021.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.
Collapse
|
117
|
Fibroblast transition to an endothelial "trans" state improves cell reprogramming efficiency. Sci Rep 2021; 11:22605. [PMID: 34799643 PMCID: PMC8604927 DOI: 10.1038/s41598-021-02056-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/02/2021] [Indexed: 01/04/2023] Open
Abstract
Fibroblast reprogramming offers the potential for myocardial regeneration via in situ cell transdifferentiation. We explored a novel strategy leveraging endothelial cell plasticity to enhance reprogramming efficiency. Rat cardiac endothelial cells and fibroblasts were treated with Gata4, Mef2c, and Tbx5 (GMT) to assess the cardio-differentiation potential of these cells. The endothelial cell transdifferentiation factor ETV2 was transiently over-expressed in fibroblasts followed by GMT treatment to assess “trans-endothelial” cardio-differentiation. Endothelial cells treated with GMT generated more cTnT+ cells than did cardiac fibroblasts (13% ± 2% vs 4% ± 0.5%, p < 0.01). Cardiac fibroblasts treated with ETV2 demonstrated increased endothelial cell markers, and when then treated with GMT yielded greater prevalence of cells expressing cardiomyocyte markers including cTnT than did fibroblasts treated with GMT or ETV2 (10.3% ± 0.2% vs 1.7% ± 0.06% and 0.6 ± 0.03, p < 0.01). Rat cardiac fibroblasts treated with GMT + ETV2 demonstrated calcium transients upon electrical stimulation and contractility synchronous with surrounding neonatal cardiomyocytes, whereas cells treated with GMT or ETV2 alone failed to contract in co-culture experiments. Human cardiac fibroblasts treated with ETV2 and then GMT likewise demonstrated greater prevalence of cTnT expression than did cells treated with GMT alone (2.8-fold increase, p < 0.05). Cardiac fibroblast transitioning through a trans-endothelial state appears to enhance cardio-differentiation by enhancing fibroblast plasticity.
Collapse
|
118
|
Haase K, Piatti F, Marcano M, Shin Y, Visone R, Redaelli A, Rasponi M, Kamm RD. Physiologic flow-conditioning limits vascular dysfunction in engineered human capillaries. Biomaterials 2021; 280:121248. [PMID: 34794827 DOI: 10.1016/j.biomaterials.2021.121248] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 02/02/2023]
Abstract
Hemodynamics play a central role in the health and disease of the coronary and peripheral vascular systems. Vessel-lining endothelial cells are known mechanosensors, responding to disturbances in flow - with mechanosensitivity hypothesized to change in response to metabolic demands. The health of our smallest microvessels have been lauded as a prognostic marker for cardiovascular health. Yet, despite numerous animal models, studying these small vessels has proved difficult. Microfluidic technologies have allowed a number of 3D vascular models to be developed and used to investigate human vessels. Here, two such systems are employed for examining 1) interstitial flow effects on neo-vessel formation, and 2) the effects of flow-conditioning on vascular remodeling following sustained static culture. Interstitial flow is shown to enhance early vessel formation via significant remodeling of vessels and interconnected tight junctions of the endothelium. In formed vessels, continuous flow maintains a stable vascular diameter and causes significant remodeling, contrasting the continued anti-angiogenic decline of statically cultured vessels. This study is the first to couple complex 3D computational flow distributions and microvessel remodeling from microvessels grown on-chip (exposed to flow or no-flow conditions). Flow-conditioned vessels (WSS < 1Pa for 30 μm vessels) increase endothelial barrier function, result in significant changes in gene expression and reduce reactive oxygen species and anti-angiogenic cytokines. Taken together, these results demonstrate microvessel mechanosensitivity to flow-conditioning, which limits deleterious vessel regression in vitro, and could have implications for future modeling of reperfusion/no-flow conditions.
Collapse
Affiliation(s)
- Kristina Haase
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Filippo Piatti
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | | | - Yoojin Shin
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Roberta Visone
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marco Rasponi
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Roger D Kamm
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA; Dept. of Biological Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
119
|
Jones BC, Shibuya S, Durkin N, De Coppi P. Regenerative medicine for childhood gastrointestinal diseases. Best Pract Res Clin Gastroenterol 2021; 56-57:101769. [PMID: 35331401 DOI: 10.1016/j.bpg.2021.101769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 01/31/2023]
Abstract
Several paediatric gastrointestinal diseases result in life-shortening organ failure. For many of these conditions, current therapeutic options are suboptimal and may not offer a cure. Regenerative medicine is an inter-disciplinary field involving biologists, engineers, and clinicians that aims to produce cell and tissue-based therapies to overcome organ failure. Exciting advances in stem cell biology, materials science, and bioengineering bring engineered gastrointestinal cell and tissue therapies to the verge of clinical trial. In this review, we summarise the requirements for bioengineered therapies, the possible sources of the various cellular and non-cellular components, and the progress towards clinical translation of oesophageal and intestinal tissue engineering to date.
Collapse
Affiliation(s)
- Brendan C Jones
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom
| | - Soichi Shibuya
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalie Durkin
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom.
| |
Collapse
|
120
|
Sugimoto S, Sato T. Organoid vs In Vivo Mouse Model: Which is Better Research Tool to Understand the Biologic Mechanisms of Intestinal Epithelium? Cell Mol Gastroenterol Hepatol 2021; 13:195-197. [PMID: 34644539 PMCID: PMC8593653 DOI: 10.1016/j.jcmgh.2021.06.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Shinya Sugimoto
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan,Correspondence Address correspondence to: Toshiro Sato, MD, PhD, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
121
|
Szklanny AA, Machour M, Redenski I, Chochola V, Goldfracht I, Kaplan B, Epshtein M, Simaan Yameen H, Merdler U, Feinberg A, Seliktar D, Korin N, Jaroš J, Levenberg S. 3D Bioprinting of Engineered Tissue Flaps with Hierarchical Vessel Networks (VesselNet) for Direct Host-To-Implant Perfusion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102661. [PMID: 34510579 PMCID: PMC11468543 DOI: 10.1002/adma.202102661] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Indexed: 05/09/2023]
Abstract
Engineering hierarchical vasculatures is critical for creating implantable functional thick tissues. Current approaches focus on fabricating mesoscale vessels for implantation or hierarchical microvascular in vitro models, but a combined approach is yet to be achieved to create engineered tissue flaps. Here, millimetric vessel-like scaffolds and 3D bioprinted vascularized tissues interconnect, creating fully engineered hierarchical vascular constructs for implantation. Endothelial and support cells spontaneously form microvascular networks in bioprinted tissues using a human collagen bioink. Sacrificial molds are used to create polymeric vessel-like scaffolds and endothelial cells seeded in their lumen form native-like endothelia. Assembling endothelialized scaffolds within vascularizing hydrogels incites the bioprinted vasculature and endothelium to cooperatively create vessels, enabling tissue perfusion through the scaffold lumen. Using a cuffing microsurgery approach, the engineered tissue is directly anastomosed with a rat femoral artery, promoting a rich host vasculature within the implanted tissue. After two weeks in vivo, contrast microcomputer tomography imaging and lectin perfusion of explanted engineered tissues verify the host ingrowth vasculature's functionality. Furthermore, the hierarchical vessel network (VesselNet) supports in vitro functionality of cardiomyocytes. Finally, the proposed approach is expanded to mimic complex structures with native-like millimetric vessels. This work presents a novel strategy aiming to create fully-engineered patient-specific thick tissue flaps.
Collapse
Affiliation(s)
- Ariel A. Szklanny
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Majd Machour
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Idan Redenski
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Václav Chochola
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrno625 00Czech Republic
| | - Idit Goldfracht
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Ben Kaplan
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Mark Epshtein
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Haneen Simaan Yameen
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Uri Merdler
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Adam Feinberg
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Dror Seliktar
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Netanel Korin
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Josef Jaroš
- Cell and Tissue RegenerationInternational Clinical Research CenterSt. Anne's University Hospital BrnoBrno65691Czech Republic
| | - Shulamit Levenberg
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| |
Collapse
|
122
|
Wingo M, Rafii S. Endothelial reprogramming for vascular regeneration: Past milestones and future directions. Semin Cell Dev Biol 2021; 122:50-55. [PMID: 34548212 DOI: 10.1016/j.semcdb.2021.09.003] [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: 06/14/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Endothelial cells are critical mediators of health and disease. Regenerative medicine techniques that target the endothelium hold vast promise for improving lifespan and quality of life worldwide. Regenerative therapies via induced pluripotent stem cells (IPSCs) have helped demonstrate disease mechanisms, but so far, concerns regarding their function, malignant potential, and expense have limited therapeutic potential. One alternative approach is direct reprogramming of somatic cells, which avoids the pluripotent state and allows for in vivo reprogramming. Transcription factors from endothelial development have yielded essential transcription factors and small molecules that induce endothelial cell fate. Most direct cell reprogramming strategies targeting endothelial cells use ETV2, a pioneer transcription factor to specify endothelial lineage via histone-modifying enzymes. Many different types of starting cells and strategies, including lentiviral transduction, inducing innate immunity, and small molecule signaling have been leveraged for reprogramming. However, so far therapeutic benefit of these strategies remains unproven. Future research will have to solve scalability, safety, and efficacy hurdles before being ready for the clinic. However, researchers have already discovered meaningful insights into disease mechanisms and development through direct reprogramming.
Collapse
Affiliation(s)
- Matthew Wingo
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
123
|
Huang K, Castiaux A, Podicheti R, Rusch DB, Martin RS, Baker LA. A Hybrid Nanofiber/Paper Cell Culture Platform for Building a 3D Blood-brain Barrier Model. SMALL METHODS 2021; 5:2100592. [PMID: 34541301 PMCID: PMC8445000 DOI: 10.1002/smtd.202100592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 05/16/2023]
Abstract
The blood brain barrier (BBB) protects the central nervous system from toxins and pathogens in the blood by regulating permeation of molecules through the barrier interface. In vitro BBB models described to date reproduce some aspects of BBB functionality, but also suffer from incomplete phenotypic expression of brain endothelial traits, difficulty in reproducibility and fabrication, or overall cost. To address these limitations, we describe a three-dimensional (3D) BBB model based on a hybrid paper/nanofiber scaffold. The cell culture platform utilizes lens paper as a framework to accommodate 3D culture of astrocytes. An electrospun nanofiber layer is coated onto one face of the paper to mimic the basement membrane and support growth of an organized two-dimensional layer of endothelial cells (ECs). Human induced pluripotent stem cell-derived ECs and astrocytes are co-cultured to develop a human BBB model. Morphological and spatial organization of model are validated with confocal microscopy. Measurements of transendothelial resistance and permeability demonstrate the BBB model develops a high-quality barrier and responds to hyperosmolar treatments. RNA-sequencing shows introduction of astrocytes both regulates EC tight junction proteins and improves endothelial phenotypes related to vasculogenesis. This model shows promise as a model platform for future in vitro studies of the BBB.
Collapse
Affiliation(s)
- Kaixiang Huang
- Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, USA
| | - Andre Castiaux
- Department of Chemistry and Center for Additive Manufacturing, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University Bloomington, 1001 East Third St., Bloomington, Indiana 47405, USA
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University Bloomington, 1001 East Third St., Bloomington, Indiana 47405, USA
| | - R Scott Martin
- Department of Chemistry and Center for Additive Manufacturing, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, USA
| | - Lane A Baker
- Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, USA
| |
Collapse
|
124
|
Dight J, Zhao J, Styke C, Khosrotehrani K, Patel J. Resident vascular endothelial progenitor definition and function: the age of reckoning. Angiogenesis 2021; 25:15-33. [PMID: 34499264 PMCID: PMC8813834 DOI: 10.1007/s10456-021-09817-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/05/2021] [Indexed: 02/07/2023]
Abstract
The cardiovascular system is composed around the central function of the endothelium that lines the inner surfaces of its vessels. In recent years, the existence of a progenitor population within the endothelium has been validated through the study of endothelial colony-forming cells (ECFCs) in human peripheral blood and certain vascular beds. However, our knowledge on endothelial populations in vivo that can give rise to ECFCs in culture has been limited. In this review we report and analyse recent attempts at describing progenitor populations in vivo from murine studies that reflect the self-renewal and stemness capacity observed in ECFCs. We pinpoint seminal discoveries within the field, which have phenotypically defined, and functionally scrutinised these endothelial progenitors. Furthermore, we review recent publications utilising single-cell sequencing technologies to better understand the endothelium in homeostasis and pathology.
Collapse
Affiliation(s)
- James Dight
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Jilai Zhao
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Cassandra Styke
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| | - Jatin Patel
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia. .,Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| |
Collapse
|
125
|
Vella G, Guelfi S, Bergers G. High Endothelial Venules: A Vascular Perspective on Tertiary Lymphoid Structures in Cancer. Front Immunol 2021; 12:736670. [PMID: 34484246 PMCID: PMC8416033 DOI: 10.3389/fimmu.2021.736670] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023] Open
Abstract
High endothelial venules (HEVs) are specialized postcapillary venules composed of cuboidal blood endothelial cells that express high levels of sulfated sialomucins to bind L-Selectin/CD62L on lymphocytes, thereby facilitating their transmigration from the blood into the lymph nodes (LN) and other secondary lymphoid organs (SLO). HEVs have also been identified in human and murine tumors in predominantly CD3+T cell-enriched areas with fewer CD20+B-cell aggregates that are reminiscent of tertiary lymphoid-like structures (TLS). While HEV/TLS areas in human tumors are predominantly associated with increased survival, tumoral HEVs (TU-HEV) in mice have shown to foster lymphocyte-enriched immune centers and boost an immune response combined with different immunotherapies. Here, we discuss the current insight into TU-HEV formation, function, and regulation in tumors and elaborate on the functional implication, opportunities, and challenges of TU-HEV formation for cancer immunotherapy.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sophie Guelfi
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Neurological Surgery, UCSF Comprehensive Cancer Center, University of California San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
126
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
127
|
Endothelial Heterogeneity in Development and Wound Healing. Cells 2021; 10:cells10092338. [PMID: 34571987 PMCID: PMC8469713 DOI: 10.3390/cells10092338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature is comprised of endothelial cells that are heterogeneous in nature. From tissue resident progenitors to mature differentiated endothelial cells, the diversity of these populations allows for the formation, maintenance, and regeneration of the vascular system in development and disease, particularly during situations of wound healing. Additionally, the de-differentiation and plasticity of different endothelial cells, especially their capacity to undergo endothelial to mesenchymal transition, has also garnered significant interest due to its implication in disease progression, with emphasis on scarring and fibrosis. In this review, we will pinpoint the seminal discoveries defining the phenotype and mechanisms of endothelial heterogeneity in development and disease, with a specific focus only on wound healing.
Collapse
|
128
|
Ryan AR, England AR, Chaney CP, Cowdin MA, Hiltabidle M, Daniel E, Gupta AK, Oxburgh L, Carroll TJ, Cleaver O. Vascular deficiencies in renal organoids and ex vivo kidney organogenesis. Dev Biol 2021; 477:98-116. [PMID: 34000274 PMCID: PMC8382085 DOI: 10.1016/j.ydbio.2021.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
Chronic kidney disease (CKD) and end stage renal disease (ESRD) are increasingly frequent and devastating conditions that have driven a surge in the need for kidney transplantation. A stark shortage of organs has fueled interest in generating viable replacement tissues ex vivo for transplantation. One promising approach has been self-organizing organoids, which mimic developmental processes and yield multicellular, organ-specific tissues. However, a recognized roadblock to this approach is that many organoid cell types fail to acquire full maturity and function. Here, we comprehensively assess the vasculature in two distinct kidney organoid models as well as in explanted embryonic kidneys. Using a variety of methods, we show that while organoids can develop a wide range of kidney cell types, as previously shown, endothelial cells (ECs) initially arise but then rapidly regress over time in culture. Vasculature of cultured embryonic kidneys exhibit similar regression. By contrast, engraftment of kidney organoids under the kidney capsule results in the formation of a stable, perfused vasculature that integrates into the organoid. This work demonstrates that kidney organoids offer a promising model system to define the complexities of vascular-nephron interactions, but the establishment and maintenance of a vascular network present unique challenges when grown ex vivo.
Collapse
Affiliation(s)
- Anne R Ryan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alicia R England
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher P Chaney
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mitzy A Cowdin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Max Hiltabidle
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward Daniel
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Thomas J Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
129
|
Tan ML, Ling L, Fischbach C. Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro. Adv Drug Deliv Rev 2021; 176:113852. [PMID: 34197895 PMCID: PMC8440401 DOI: 10.1016/j.addr.2021.113852] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Despite decades of research and advancements in diagnostic and treatment modalities, cancer remains a major global healthcare challenge. This is due in part to a lack of model systems that allow investigating the mechanisms underlying tumor development, progression, and therapy resistance under relevant conditions in vitro. Tumor cell interactions with their surroundings influence all stages of tumorigenesis and are shaped by both biological and biophysical cues including cell-cell and cell-extracellular matrix (ECM) interactions, tissue architecture and mechanics, and mass transport. Engineered tumor models provide promising platforms to elucidate the individual and combined contributions of these cues to tumor malignancy under controlled and physiologically relevant conditions. This review will summarize current knowledge of the biological and biophysical microenvironmental cues that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions and highlight advancements in tumor engineering approaches to further improve these technologies.
Collapse
Affiliation(s)
- Matthew L Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
130
|
Cell-based therapies for vascular regeneration: Past, present and future. Pharmacol Ther 2021; 231:107976. [PMID: 34480961 DOI: 10.1016/j.pharmthera.2021.107976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/01/2021] [Accepted: 08/05/2021] [Indexed: 12/27/2022]
Abstract
Tissue vascularization remains one of the outstanding challenges in regenerative medicine. Beyond its role in circulating oxygen and nutrients, the vasculature is critical for organ development, function and homeostasis. Importantly, effective vascular regeneration is key in generating large 3D tissues for regenerative medicine applications to enable the survival of cells post-transplantation, organ growth, and integration into the host system. Therefore, the absence of clinically applicable means of (re)generating vessels is one of the main obstacles in cell replacement therapy. In this review, we highlight cell-based vascularization strategies which demonstrate clinical potential, discuss their strengths and limitations and highlight the main obstacles hindering cell-based therapeutic vascularization.
Collapse
|
131
|
Ahn Y, An JH, Yang HJ, Lee DG, Kim J, Koh H, Park YH, Song BS, Sim BW, Lee HJ, Lee JH, Kim SU. Human Blood Vessel Organoids Penetrate Human Cerebral Organoids and Form a Vessel-Like System. Cells 2021; 10:cells10082036. [PMID: 34440805 PMCID: PMC8393185 DOI: 10.3390/cells10082036] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 01/19/2023] Open
Abstract
Vascularization of tissues, organoids and organ-on-chip models has been attempted using endothelial cells. However, the cultured endothelial cells lack the capacity to interact with other somatic cell types, which is distinct from developing vascular cells in vivo. Recently, it was demonstrated that blood vessel organoids (BVOs) recreate the structure and functions of developing human blood vessels. However, the tissue-specific adaptability of BVOs had not been assessed in somatic tissues. Herein, we investigated whether BVOs infiltrate human cerebral organoids and form a blood-brain barrier. As a result, vascular cells arising from BVOs penetrated the cerebral organoids and developed a vessel-like architecture composed of CD31+ endothelial tubes coated with SMA+ or PDGFR+ mural cells. Molecular markers of the blood-brain barrier were detected in the vascularized cerebral organoids. We revealed that BVOs can form neural-specific blood-vessel networks that can be maintained for over 50 days.
Collapse
Affiliation(s)
- Yujin Ahn
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Ju-Hyun An
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Hae-Jun Yang
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
| | - Dong Gil Lee
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
| | - Jieun Kim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyebin Koh
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Young-Ho Park
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
| | - Bong-Seok Song
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
| | - Bo-Woong Sim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
| | - Hong J. Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea;
- Research Institute, eBiogen Inc., Seoul 04785, Korea
| | - Jong-Hee Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea
- Correspondence: (J.-H.L.); (S.-U.K.); Tel.: +82-43-240-6312 (J.-H.L.); +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Sun-Uk Kim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Korea; (Y.A.); (J.-H.A.); (H.-J.Y.); (D.G.L.); (J.K.); (H.K.); (Y.-H.P.); (B.-S.S.); (B.-W.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (J.-H.L.); (S.-U.K.); Tel.: +82-43-240-6312 (J.-H.L.); +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| |
Collapse
|
132
|
Evans P, Wojta J, Hoefer IE, Waltenberger J, Guzik T, Badimon L, Weber C. The year in basic vascular biology research: from mechanoreceptors and neutrophil extracellular traps to smartphone data and omics. Cardiovasc Res 2021; 117:1814-1822. [PMID: 33744925 PMCID: PMC8083796 DOI: 10.1093/cvr/cvab105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
2020 has been an extraordinary year. The emergence of COVID-19 has driven urgent research in pulmonary and cardiovascular science and other fields. It has also shaped the way that we work with many experimental laboratories shutting down for several months, while bioinformatics approaches and other large data projects have gained prominence. Despite these setbacks, vascular biology research is stronger than ever. On behalf of the European Society of Cardiology Council for Basic Cardiovascular Science (ESC CBCS), here we review some of the vascular biology research highlights for 2020. This review is not exhaustive and there are many outstanding vascular biology publications that we were unable to cite due to page limits. Notwithstanding this, we have provided a snapshot of vascular biology research excellence in 2020 and identify topics that are in the ascendency and likely to gain prominence in coming years.
Collapse
Affiliation(s)
- Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.,Insigneo Institute, Sheffield, UK
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Imo E Hoefer
- Central Diagnostic Laboratory, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Department of Medicine, Jagiellonian University Collegium Medicum, Cracow, Poland
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian-Universität (LMU) München, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; and.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
133
|
Choudhuri A, Han T, Zon LI. From development toward therapeutics, a collaborative effort on blood progenitors. Stem Cell Reports 2021; 16:1674-1685. [PMID: 34115985 PMCID: PMC8486953 DOI: 10.1016/j.stemcr.2021.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/02/2023] Open
Abstract
The National Heart, Lung, and Blood Institute Progenitor Cell Translational Consortium Blood Progenitor Meeting was hosted virtually on November 5, 2020, with 93 attendees across 20 research groups. The purpose of this meeting was to exchange recent findings, discuss current efforts, and identify prospective opportunities in the field of hematopoietic stem and progenitor cell research and therapeutic discovery.
Collapse
Affiliation(s)
- Avik Choudhuri
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Children's Hospital Boston, Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Tianxiao Han
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Children's Hospital Boston, Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Children's Hospital Boston, Howard Hughes Medical Institute, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Stem Cell Institute, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
134
|
Adelson RP, Palikuqi B, Weiss Z, Checco A, Schreiner R, Rafii S, Rabbany SY. Morphological characterization of Etv2 vascular explants using fractal analysis and atomic force microscopy. Microvasc Res 2021; 138:104205. [PMID: 34146583 DOI: 10.1016/j.mvr.2021.104205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 11/25/2022]
Abstract
The rapid engraftment of vascular networks is critical for functional incorporation of tissue explants. However, existing methods for inducing angiogenesis utilize approaches that yield vasculature with poor temporal stability or inadequate mechanical integrity, which reduce their robustness in vivo. The transcription factor Ets variant 2 (Etv2) specifies embryonic hematopoietic and vascular endothelial cell (EC) development, and is transiently reactivated during postnatal vascular regeneration and tumor angiogenesis. This study investigates the role for Etv2 upregulation in forming stable vascular beds both in vitro and in vivo. Control and Etv2+ prototypical fetal-derived human umbilical vein ECs (HUVECs) and adult ECs were angiogenically grown into vascular beds. These vessel beds were characterized using fractal dimension and lacunarity, to quantify their branching complexity and space-filling homogeneity, respectively. Atomic force microscopy (AFM) was used to explore whether greater complexity and homogeneity lead to more mechanically stable vessels. Additionally, markers of EC integrity were used to probe for mechanistic clues. Etv2+ HUVECs exhibit greater branching, vessel density, and structural homogeneity, and decreased stiffness in vitro and in vivo, indicating a greater propensity for stable vessel formation. When co-cultured with colon tumor organoid tissue, Etv2+ HUVECs had decreased fractal dimension and lacunarity compared to Etv2+ HUVECs cultured alone, indicating that vessel density and homogeneity of vessel spacing increased due to the presence of Etv2. This study sets forth the novel concept that fractal dimension, lacunarity, and AFM are as informative as conventional angiogenic measurements, including vessel branching and density, to assess vascular perfusion and stability.
Collapse
Affiliation(s)
- Robert P Adelson
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Brisa Palikuqi
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zachary Weiss
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Antonio Checco
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Ryan Schreiner
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sina Y Rabbany
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA; Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
135
|
Mandrycky CJ, Howard CC, Rayner SG, Shin YJ, Zheng Y. Organ-on-a-chip systems for vascular biology. J Mol Cell Cardiol 2021; 159:1-13. [PMID: 34118217 DOI: 10.1016/j.yjmcc.2021.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/03/2021] [Accepted: 06/06/2021] [Indexed: 12/18/2022]
Abstract
Organ-on-a-chip (OOC) platforms involve the miniaturization of cell culture systems and enable a variety of novel experimental approaches. These range from modeling the independent effects of biophysical forces on cells to screening novel drugs in multi-organ microphysiological systems, all within microscale devices. As in living systems, the incorporation of vascular structure is a key feature common to almost all organ-on-a-chip systems. In this review we highlight recent advances in organ-on-a-chip technologies with a focus on the vasculature. We first present the developmental process of the blood vessels through which vascular cells assemble into networks and remodel to form complex vascular beds under flow. We then review self-assembled vascular models and flow systems for the study of vascular development and biology as well as pre-patterned vascular models for the generation of perfusable microvessels for modeling vascular and tissue function. We finally conclude with a perspective on developing future OOC approaches for studying different aspects of vascular biology. We highlight the fit for purpose selection of OOC models towards either simple but powerful testbeds for therapeutic development, or complex vasculature to accurately replicate human physiology for specific disease modeling and tissue regeneration.
Collapse
Affiliation(s)
- Christian J Mandrycky
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Caitlin C Howard
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Samuel G Rayner
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA; Department of Medicine; Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Yu Jung Shin
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Ying Zheng
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
136
|
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: 8.0] [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
|
137
|
Zhang Q, Bosch-Rué È, Pérez RA, Truskey GA. Biofabrication of tissue engineering vascular systems. APL Bioeng 2021; 5:021507. [PMID: 33981941 PMCID: PMC8106537 DOI: 10.1063/5.0039628] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death among persons aged 65 and older in the United States and many other developed countries. Tissue engineered vascular systems (TEVS) can serve as grafts for CVD treatment and be used as in vitro model systems to examine the role of various genetic factors during the CVD progressions. Current focus in the field is to fabricate TEVS that more closely resembles the mechanical properties and extracellular matrix environment of native vessels, which depends heavily on the advance in biofabrication techniques and discovery of novel biomaterials. In this review, we outline the mechanical and biological design requirements of TEVS and explore the history and recent advances in biofabrication methods and biomaterials for tissue engineered blood vessels and microvascular systems with special focus on in vitro applications. In vitro applications of TEVS for disease modeling are discussed.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Èlia Bosch-Rué
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès 08195, Spain
| | - Román A. Pérez
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès 08195, Spain
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
138
|
Abbas M, Moradi F, Hu W, Regudo KL, Osborne M, Pettipas J, Atallah DS, Hachem R, Ott-Peron N, Stuart JA. Vertebrate cell culture as an experimental approach – limitations and solutions. Comp Biochem Physiol B Biochem Mol Biol 2021; 254:110570. [DOI: 10.1016/j.cbpb.2021.110570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
|
139
|
Marsee A, Roos FJM, Verstegen MMA, Gehart H, de Koning E, Lemaigre F, Forbes SJ, Peng WC, Huch M, Takebe T, Vallier L, Clevers H, van der Laan LJW, Spee B. Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids. Cell Stem Cell 2021; 28:816-832. [PMID: 33961769 PMCID: PMC11699540 DOI: 10.1016/j.stem.2021.04.005] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatic, pancreatic, and biliary (HPB) organoids are powerful tools for studying development, disease, and regeneration. As organoid research expands, the need for clear definitions and nomenclature describing these systems also grows. To facilitate scientific communication and consistent interpretation, we revisit the concept of an organoid and introduce an intuitive classification system and nomenclature for describing these 3D structures through the consensus of experts in the field. To promote the standardization and validation of HPB organoids, we propose guidelines for establishing, characterizing, and benchmarking future systems. Finally, we address some of the major challenges to the clinical application of organoids.
Collapse
Affiliation(s)
- Ary Marsee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Floris J M Roos
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Helmuth Gehart
- Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Eelco de Koning
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands; Leiden University Medical Center, Department of Medicine, Leiden, the Netherlands
| | - Frédéric Lemaigre
- Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Stuart J Forbes
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Weng Chuan Peng
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, and Center for Stem Cell, and Organoid Medicine (CuSTOM), Cincinnati Children Hospital Medical Center, Cincinnati, OH, USA; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, Cambridgeshire, UK; Department of Surgery, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Center, Cambridge, UK
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
140
|
Primate Organoids and Gene-Editing Technologies toward Next-Generation Biomedical Research. Trends Biotechnol 2021; 39:1332-1342. [PMID: 33941418 DOI: 10.1016/j.tibtech.2021.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/08/2023]
Abstract
The improved ability to organize pluripotent stem cells (PSCs) into 3D structures in vitro has shed light on organoid technology to recapitulate organs and tumors in vivo. Advances in gene-editing technologies, particularly CRISPR-mediated techniques, offer tremendous potential in facilitating organoid research, including the study of development, disease modeling, and personalized medicine. This review discusses how the combination of two novel technologies - organoids and gene editing - not only contributes to revealing molecular events taking place during development and tumorigenesis but also has implications for biobanking, precision medicine, and other diverse biomedical applications.
Collapse
|
141
|
Bessy T, Itkin T, Passaro D. Bioengineering the Bone Marrow Vascular Niche. Front Cell Dev Biol 2021; 9:645496. [PMID: 33996805 PMCID: PMC8113773 DOI: 10.3389/fcell.2021.645496] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/23/2021] [Indexed: 01/01/2023] Open
Abstract
The bone marrow (BM) tissue is the main physiological site for adult hematopoiesis. In recent years, the cellular and matrix components composing the BM have been defined with unprecedent resolution, both at the molecular and structural levels. With the expansion of this knowledge, the possibility of reproducing a BM-like structure, to ectopically support and study hematopoiesis, becomes a reality. A number of experimental systems have been implemented and have displayed the feasibility of bioengineering BM tissues, supported by cells of mesenchymal origin. Despite being known as an abundant component of the BM, the vasculature has been largely disregarded for its role in regulating tissue formation, organization and determination. Recent reports have highlighted the crucial role for vascular endothelial cells in shaping tissue development and supporting steady state, emergency and malignant hematopoiesis, both pre- and postnatally. Herein, we review the field of BM-tissue bioengineering with a particular focus on vascular system implementation and integration, starting from describing a variety of applicable in vitro models, ending up with in vivo preclinical models. Additionally, we highlight the challenges of the field and discuss the clinical perspectives in terms of adoptive transfer of vascularized BM-niche grafts in patients to support recovering hematopoiesis.
Collapse
Affiliation(s)
- Thomas Bessy
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| | - Tomer Itkin
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Diana Passaro
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
142
|
Gel-Free 3D Tumoroids with Stem Cell Properties Modeling Drug Resistance to Cisplatin and Imatinib in Metastatic Colorectal Cancer. Cells 2021; 10:cells10020344. [PMID: 33562088 PMCID: PMC7914642 DOI: 10.3390/cells10020344] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 01/16/2023] Open
Abstract
Researchers have developed several three-dimensional (3D) culture systems, including spheroids, organoids, and tumoroids with increased properties of cancer stem cells (CSCs), also called cancer-initiating cells (CICs). Drug resistance is a crucial issue involving recurrence in cancer patients. Many studies on anti-cancer drugs have been reported using 2D culture systems, whereas 3D cultured tumoroids have many advantages for assessing drug sensitivity and resistance. Here, we aimed to investigate whether Cisplatin (a DNA crosslinker), Imatinib (a multiple tyrosine kinase inhibitor), and 5-Fluorouracil (5-FU: an antimetabolite) alter the tumoroid growth of metastatic colorectal cancer (mCRC). Gene expression signatures of highly metastatic aggregative CRC (LuM1 cells) vs. low-metastatic, non-aggregative CRC (Colon26 and NM11 cells) were analyzed using microarray. To establish a 3D culture-based multiplexing reporter assay system, LuM1 was stably transfected with the Mmp9 promoter-driven ZsGreen fluorescence reporter gene, which was designated as LuM1/m9 cells and cultured in NanoCulture Plate®, a gel-free 3D culture device. LuM1 cells highly expressed mRNA encoding ABCG2 (a drug resistance pump, i.e., CSC/CIC marker), other CSC/CIC markers (DLL1, EpCAM, podoplanin, STAT3/5), pluripotent stem cell markers (Sox4/7, N-myc, GATA3, Nanog), and metastatic markers (MMPs, Integrins, EGFR), compared to the other two cell types. Hoechst efflux stem cell-like side population was increased in LuM1 (7.8%) compared with Colon26 (2.9%), both of which were markedly reduced by verapamil treatment, an ABCG2 inhibitor. Smaller cell aggregates of LuM1 were more sensitive to Cisplatin (at 10 μM), whereas larger tumoroids with increased ABCG2 expression were insensitive. Notably, Cisplatin (2 μM) and Imatinib (10 μM) at low concentrations significantly promoted tumoroid formation (cell aggregation) and increased Mmp9 promoter activity in mCRC LuM1/m9, while not cytotoxic to them. On the other hand, 5-FU significantly inhibited tumoroid growth, although not completely. Thus, drug resistance in cancer with increased stem cell properties was modeled using the gel-free 3D cultured tumoroid system. The tumoroid culture is useful and easily accessible for the assessment of drug sensitivity and resistance.
Collapse
|
143
|
De Coppi P, Grikscheit TC. Regeneration and tissue engineering: How pediatric surgeons contributed to building a new field to change the future of medicine. Semin Pediatr Surg 2021; 30:151018. [PMID: 33648705 DOI: 10.1016/j.sempedsurg.2021.151018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The authors highlight the speciality field of regenerative medicine and its application to health care. Academic pediatric surgeons have been the early pioneers here sharing exciting discovery and the opportunities for research enterprise. An overview of current and future therapeutics is provided for the reader.
Collapse
Affiliation(s)
- Paolo De Coppi
- Surgery Unit, Great Ormond Street Institute of Child Health, University College London, Great Ormond St. Hospital for Children, 30 Guilford St., London WC1N 1EH, United Kingdom.
| | - T C Grikscheit
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital, Los Angeles, CA, United States of America
| |
Collapse
|
144
|
Zhang T, Liu D, Wang Y, Sun M, Xia L. The E-Twenty-Six Family in Hepatocellular Carcinoma: Moving into the Spotlight. Front Oncol 2021; 10:620352. [PMID: 33585247 PMCID: PMC7873604 DOI: 10.3389/fonc.2020.620352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/08/2020] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of morbidity and mortality worldwide. Although therapeutic strategies have recently advanced, tumor metastasis and drug resistance continue to pose challenges in the treatment of HCC. Therefore, new molecular targets are needed to develop novel therapeutic strategies for this cancer. E-twenty-six (ETS) transcription family has been implicated in human malignancies pathogenesis and progression, including leukemia, Ewing sarcoma, gastrointestinal stromal tumors. Recently, increasing studies have expanded its great potential as functional players in other cancers, including HCC. This review focuses primarily on the key functions and molecular mechanisms of ETS factors in HCC. Elucidating these molecular details may provide novel potential therapeutic strategies for cancers.
Collapse
Affiliation(s)
| | | | | | | | - Limin Xia
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
145
|
A Perfusion Bioreactor for Longitudinal Monitoring of Bioengineered Liver Constructs. NANOMATERIALS 2021; 11:nano11020275. [PMID: 33494337 PMCID: PMC7912543 DOI: 10.3390/nano11020275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
In the field of in vitro liver disease models, decellularised organ scaffolds maintain the original biomechanical and biological properties of the extracellular matrix and are established supports for in vitro cell culture. However, tissue engineering approaches based on whole organ decellularized scaffolds are hampered by the scarcity of appropriate bioreactors that provide controlled 3D culture conditions. Novel specific bioreactors are needed to support long-term culture of bioengineered constructs allowing non-invasive longitudinal monitoring. Here, we designed and validated a specific bioreactor for long-term 3D culture of whole liver constructs. Whole liver scaffolds were generated by perfusion decellularisation of rat livers. Scaffolds were seeded with Luc+HepG2 and primary human hepatocytes and cultured in static or dynamic conditions using the custom-made bioreactor. The bioreactor included a syringe pump, for continuous unidirectional flow, and a circuit built to allow non-invasive monitoring of culture parameters and media sampling. The bioreactor allowed non-invasive analysis of cell viability, distribution, and function of Luc+HepG2-bioengineered livers cultured for up to 11 days. Constructs cultured in dynamic conditions in the bioreactor showed significantly higher cell viability, measured with bioluminescence, distribution, and functionality (determined by albumin production and expression of CYP enzymes) in comparison to static culture conditions. Finally, our bioreactor supports primary human hepatocyte viability and function for up to 30 days, when seeded in the whole liver scaffolds. Overall, our novel bioreactor is capable of supporting cell survival and metabolism and is suitable for liver tissue engineering for the development of 3D liver disease models.
Collapse
|
146
|
YAP and endothelin-1 signaling: an emerging alliance in cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:27. [PMID: 33422090 PMCID: PMC7797087 DOI: 10.1186/s13046-021-01827-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/03/2021] [Indexed: 12/14/2022]
Abstract
The rational making the G protein-coupled receptors (GPCR) the centerpiece of targeted therapies is fueled by the awareness that GPCR-initiated signaling acts as pivotal driver of the early stages of progression in a broad landscape of human malignancies. The endothelin-1 (ET-1) receptors (ET-1R), known as ETA receptor (ETAR) and ETB receptor (ETBR) that belong to the GPCR superfamily, affect both cancer initiation and progression in a variety of cancer types. By the cross-talking with multiple signaling pathways mainly through the scaffold protein β-arrestin1 (β-arr1), ET-1R axis cooperates with an array of molecular determinants, including transcription factors and co-factors, strongly affecting tumor cell fate and behavior. In this scenario, recent findings shed light on the interplay between ET-1 and the Hippo pathway. In ETAR highly expressing tumors ET-1 axis induces the de-phosphorylation and nuclear accumulation of the Hippo pathway downstream effectors, the paralogous transcriptional cofactors Yes-associated protein (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ). Recent evidence have discovered that ET-1R/β-arr1 axis instigates a transcriptional interplay involving YAP and mutant p53 proteins, which share a common gene signature and cooperate in a oncogenic signaling network. Mechanistically, YAP and mutp53 are enrolled in nuclear complexes that turn on a highly selective YAP/mutp53-dependent transcriptional response. Notably, ET-1R blockade by the FDA approved dual ET-1 receptor antagonist macitentan interferes with ET-1R/YAP/mutp53 signaling interplay, through the simultaneous suppression of YAP and mutp53 functions, hampering metastasis and therapy resistance. Based on these evidences, we aim to review the recent findings linking the GPCR signaling, as for ET-1R, to YAP/TAZ signaling, underlining the clinical relevance of the blockade of such signaling network in the tumor and microenvironmental contexts. In particular, we debate the clinical implications regarding the use of dual ET-1R antagonists to blunt gain of function activity of mutant p53 proteins and thereby considering them as a potential therapeutic option for mutant p53 cancers. The identification of ET-1R/β-arr1-intertwined and bi-directional signaling pathways as targetable vulnerabilities, may open new therapeutic approaches able to disable the ET-1R-orchestrated YAP/mutp53 signaling network in both tumor and stromal cells and concurrently sensitizes to high-efficacy combined therapeutics.
Collapse
|
147
|
van der Valk WH, Steinhart MR, Zhang J, Koehler KR. Building inner ears: recent advances and future challenges for in vitro organoid systems. Cell Death Differ 2020; 28:24-34. [PMID: 33318601 PMCID: PMC7853146 DOI: 10.1038/s41418-020-00678-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
While inner ear disorders are common, our ability to intervene and recover their sensory function is limited. In vitro models of the inner ear, like the organoid system, could aid in identifying new regenerative drugs and gene therapies. Here, we provide a perspective on the status of in vitro inner ear models and guidance on how to improve their applicability in translational research. We highlight the generation of inner ear cell types from pluripotent stem cells as a particularly promising focus of research. Several exciting recent studies have shown how the developmental signaling cues of embryonic and fetal development can be mimicked to differentiate stem cells into “inner ear organoids” containing otic progenitor cells, hair cells, and neurons. However, current differentiation protocols and our knowledge of embryonic and fetal inner ear development in general, have a bias toward the sensory epithelia of the inner ear. We propose that a more holistic view is needed to better model the inner ear in vitro. Moving forward, attention should be made to the broader diversity of neuroglial and mesenchymal cell types of the inner ear, and how they interact in space or time during development. With improved control of epithelial, neuroglial, and mesenchymal cell fate specification, inner ear organoids would have the ability to truly recapitulate neurosensory function and dysfunction. We conclude by discussing how single-cell atlases of the developing inner ear and technical innovations will be critical tools to advance inner ear organoid platforms for future pre-clinical applications.
Collapse
Affiliation(s)
- Wouter H van der Valk
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Matthew R Steinhart
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingyuan Zhang
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Karl R Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA. .,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
148
|
Haykal MM, Nahmias C, Varon C, Martin OCB. Organotypic Modeling of the Tumor Landscape. Front Cell Dev Biol 2020; 8:606039. [PMID: 33330508 PMCID: PMC7732527 DOI: 10.3389/fcell.2020.606039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer is a complex disease and it is now clear that not only epithelial tumor cells play a role in carcinogenesis. The tumor microenvironment is composed of non-stromal cells, including endothelial cells, adipocytes, immune and nerve cells, and a stromal compartment composed of extracellular matrix, cancer-associated fibroblasts and mesenchymal cells. Tumorigenesis is a dynamic process with constant interactions occurring between the tumor cells and their surroundings. Even though all connections have not yet been discovered, it is now known that crosstalk between actors of the microenvironment drives cancer progression. Taking into account this complexity, it is important to develop relevant models to study carcinogenesis. Conventional 2D culture models fail to represent the entire tumor microenvironment properly and the use of animal models should be decreased with respect to the 3Rs rule. To this aim, in vitro organotypic models have been significantly developed these past few years. These models have different levels of complexity and allow the study of tumor cells alone or in interaction with the microenvironment actors during the multiple stages of carcinogenesis. This review depicts recent insights into organotypic modeling of the tumor and its microenvironment all throughout cancer progression. It offers an overview of the crosstalk between epithelial cancer cells and their microenvironment during the different phases of carcinogenesis, from the early cell autonomous events to the late metastatic stages. The advantages of 3D over classical 2D or in vivo models are presented as well as the most promising organotypic models. A particular focus is made on organotypic models used for studying cancer progression, from the less complex spheroids to the more sophisticated body-on-a-chip. Last but not least, we address the potential benefits of these models in personalized medicine which is undoubtedly a domain paving the path to new hopes in terms of cancer care and cure.
Collapse
Affiliation(s)
- Maria M. Haykal
- Université Paris-Saclay, Institut Gustave Roussy, Inserm U981, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | - Clara Nahmias
- Université Paris-Saclay, Institut Gustave Roussy, Inserm U981, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | | | | |
Collapse
|
149
|
Brassard-Jollive N, Monnot C, Muller L, Germain S. In vitro 3D Systems to Model Tumor Angiogenesis and Interactions With Stromal Cells. Front Cell Dev Biol 2020; 8:594903. [PMID: 33224956 PMCID: PMC7674638 DOI: 10.3389/fcell.2020.594903] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
In vitro 3D culture systems provide promising tools for screening novel therapies and understanding drug resistance mechanisms in cancer because they are adapted for high throughput analysis. One of the main current challenges is to reproducibly culture patient samples containing cancer and stromal cells to faithfully recapitulate tumor microenvironment and move toward efficient personalized medicine. Tumors are composed of heterogeneous cell populations and characterized by chaotic vascularization in a remodeled microenvironment. Indeed, tumor angiogenesis occurs in a complex stroma containing immune cells and cancer-associated fibroblasts that secrete important amounts of cytokines, growth factors, extracellular vesicles, and extracellular matrix (ECM). This process leads to the formation of inflated, tortuous, and permeable capillaries that display deficient basement membrane (BM) and perivascular coverage. These abnormal capillaries affect responses to anti-cancer therapies such as anti-angiogenic, radio-, and immunotherapies. Current pre-clinical models are limited for investigating interactions between tumor cells and vascularization during tumor progression as well as mechanisms that lead to drug resistance. In vitro approaches developed for vascularization are either the result of engineered cell lining or based on physiological processes including vasculogenesis and sprouting angiogenesis. They allow investigation of paracrine and direct interactions between endothelial and tumor and/or stromal cells, as well as impact of biochemical and biophysical cues of the microenvironment, using either natural matrix components or functionalized synthetic hydrogels. In addition, microfluidic devices provide access to modeling the impact of shear stress and interstitial flow and growth factor gradients. In this review, we will describe the state of the art co-culture models of vascularized micro-tumors in order to study tumor progression and metastatic dissemination including intravasation and/or extravasation processes.
Collapse
Affiliation(s)
- Noémie Brassard-Jollive
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, CNRS UMR 7241, INSERM U1050, PSL Research University, Paris, France
| |
Collapse
|