1
|
Liu H, Xu H, Zhu Y, Wang Z, Hu D, Yang L, Zhu Y, Galan EA, Huang R, Peng H, Ma S. A Large Model-Derived Algorithm for Complex Organoids with Internal Morphogenesis and Digital Marker Derivation. Anal Chem 2024; 96:19258-19266. [PMID: 39445667 DOI: 10.1021/acs.analchem.4c02212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Automated segmentation and evaluation algorithms have been demonstrated to enhance the simplicity and translational utility of organoid technology. However, there is a pressing need for the development of complex organoids that possess epithelium environmental elements, dense regional cell aggregation, and intraorganoid morphologies. Nevertheless, there has been limited progress, including both the construction of data sets and the development of algorithms, in the use of user-friendly microscopy to address such complex organoids. In this study, a data set of bright-field and living cell fluorescence images in paired forms and with temporal variance was constructed using droplet-engineered lung organoids. Additionally, a large model-based algorithm was developed. Both the organoid contours and intraorganoid morphologies were included in the data set, and their physical parameters were included and screened to form multiplex digital markers for organoid evaluation. The algorithm has been demonstrated to outperform existing methods and is therefore suitable for the evaluation of complex organoids. It is expected that the algorithm will facilitate the successful demonstration of AI in organoid evaluation and decision-making regarding their status.
Collapse
Affiliation(s)
- Hanghang Liu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Haohan Xu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Yu Zhu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Danni Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Lingxiao Yang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Yinheng Zhu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Edgar A Galan
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Ruqi Huang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Haiying Peng
- General Hospital of the Southern Theater Command of the Chinese People's Liberation Army, Guangzhou 510280, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
2
|
Vallmajo-Martin Q, Ma Z, Srinivasan S, Murali D, Dravis C, Mukund K, Subramaniam S, Wahl GM, Lytle NK. The molecular chronology of mammary epithelial cell fate switching. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617155. [PMID: 39415993 PMCID: PMC11482796 DOI: 10.1101/2024.10.08.617155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The adult mammary gland is maintained by lineage-restricted progenitor cells through pregnancy, lactation, involution, and menopause. Injury resolution, transplantation-associated mammary gland reconstitution, and tumorigenesis are unique exceptions, wherein mammary basal cells gain the ability to reprogram to a luminal state. Here, we leverage newly developed cell-identity reporter mouse strains, and time-resolved single-cell epigenetic and transcriptomic analyses to decipher the molecular programs underlying basal-to-luminal fate switching in vivo. We demonstrate that basal cells rapidly reprogram toward plastic cycling intermediates that appear to hijack molecular programs we find in bipotent fetal mammary stem cells and puberty-associatiated cap cells. Loss of basal-cell specifiers early in dedifferentiation coincides with activation of Notch and BMP, among others. Pharmacologic blockade of each pathway disrupts basal-to-luminal transdifferentiation. Our studies provide a comprehensive map and resource for understanding the coordinated molecular changes enabling terminally differentiated epithelial cells to transition between cell lineages and highlights the stunning rapidity by which epigenetic reprogramming can occur in response to disruption of tissue structure.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Zhibo Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Sumana Srinivasan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Divya Murali
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Christopher Dravis
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Geoffrey M. Wahl
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nikki K. Lytle
- Department of Surgery, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- These authors contributed equally
| |
Collapse
|
3
|
Ahammed B, Kalangi SK. A Decade of Organoid Research: Progress and Challenges in the Field of Organoid Technology. ACS OMEGA 2024; 9:30087-30096. [PMID: 39035960 PMCID: PMC11256333 DOI: 10.1021/acsomega.4c03683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024]
Abstract
Organoid technology, revolutionizing biomedical research, offers a transformative approach to studying human developmental biology, disease pathology, and drug discovery. Originating from the pioneering work of Henry Van Peters Wilson in 1907 and evolving through subsequent breakthroughs, organoids are three-dimensional structures derived from stem cells or tissue explants that mimic the architecture and function of organs in vitro. With the ability to model various organs such as intestine, liver, brain, kidney, and more, organoids provide unprecedented insights into organ development, disease mechanisms, and drug responses. This review highlights the historical context, generation methods, applications, and challenges of organoid technology. Furthermore, it discusses recent advancements, including strategies to address hypoxia-induced cell death and enhance vascularization within organoids, aiming to refine their physiological relevance and unlock their full potential in personalized medicine and organ transplantation.
Collapse
Affiliation(s)
- Basheer Ahammed
- West BC Colony,
Guduru, Kurnool, Andhra Pradesh 518466, India
| | - Suresh K. Kalangi
- Molecular
Microbiology and Immunology Division, CSIR—Central Drug Research
Institute, Lucknow 226031, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
4
|
Muthuswamy SK, Brugge JS. Organoid Cultures for the Study of Mammary Biology and Breast Cancer: The Promise and Challenges. Cold Spring Harb Perspect Med 2024; 14:a041661. [PMID: 38110241 PMCID: PMC11216180 DOI: 10.1101/cshperspect.a041661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
During the last decade, biomedical research has experienced a resurgence in the use of three-dimensional culture models for studies of normal and cancer biology. This resurgence has been driven by the development of models in which primary cells are grown in tissue-mimicking media and extracellular matrices to create organoid or organotypic cultures that more faithfully replicate the complex architecture and physiology of normal tissues and tumors. In addition, patient-derived tumor organoids preserve the three-dimensional organization and characteristics of the patient tumors ex vivo, becoming excellent preclinical models to supplement studies of tumor xenografts transplanted into immunocompromised mice. In this perspective, we provide an overview of how organoids are being used to investigate normal mammary biology and as preclinical models of breast cancer and discuss improvements that would enhance their utility and relevance to the field.
Collapse
Affiliation(s)
- Senthil K Muthuswamy
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland 20894, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Ludwig Center at Harvard, Harvard Medical School Boston, Boston, Massachusetts 02115, USA
| |
Collapse
|
5
|
Caruso M, Saberiseyedabad K, Mourao L, Scheele CLGJ. A Decision Tree to Guide Human and Mouse Mammary Organoid Model Selection. Methods Mol Biol 2024; 2764:77-105. [PMID: 38393590 DOI: 10.1007/978-1-0716-3674-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Over the past 50 years, researchers from the mammary gland field have launched a collection of distinctive 3D cell culture systems to study multiple aspects of mammary gland physiology and disease. As our knowledge about the mammary gland evolves, more sophisticated 3D cell culture systems are required to answer more and more complex questions. Nowadays, morphologically complex mammary organoids can be generated in distinct 3D settings, along with reproduction of multiple aspects of the gland microenvironment. Yet, each 3D culture protocol comes with its advantages and limitations, where some culture systems are best suited to study stemness potential, whereas others are tailored towards the study of mammary gland morphogenesis. Therefore, prior to starting a 3D mammary culture experiment, it is important to consider and select the ideal culture model to address the biological question of interest. The number and technical requirements of novel 3D cell culture methods vastly increased over the past decades, making it currently challenging and time consuming to identify the best experimental testing. In this chapter, we provide a summary of the most promising murine and human 3D organoid models that are currently used in mammary gland biology research. For each model, we will provide a brief description of the protocol and an overview of the expected morphological outcome, the advantages of the model, and the potential pitfalls, to guide the reader to the best model of choice for specific applications.
Collapse
Affiliation(s)
- Marika Caruso
- VIB-KU Leuven Center for Cancer Biology, Department of Oncology, Leuven, Belgium
| | | | - Larissa Mourao
- VIB-KU Leuven Center for Cancer Biology, Department of Oncology, Leuven, Belgium
| | | |
Collapse
|
6
|
Englund JI, Bui H, Dinç DD, Paavolainen O, McKenna T, Laitinen S, Munne P, Klefström J, Peuhu E, Katajisto P. Laminin matrix adhesion regulates basal mammary epithelial cell identity. J Cell Sci 2022; 135:285829. [DOI: 10.1242/jcs.260232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/28/2022] [Indexed: 12/07/2022] Open
Abstract
ABSTRACT
Mammary epithelium is a bilayered ductal network composed of luminal and basal epithelial cells, which together drive the growth and functional differentiation of the gland. Basal mammary epithelial cells (MECs) exhibit remarkable plasticity and progenitor activity that facilitate epithelial expansion. However, their activity must be tightly regulated to restrict excess basal cell activity. Here, we show that adhesion of basal cells to laminin α5-containing basement membrane matrix, which is produced by luminal cells, presents such a control mechanism. Adhesion to laminin α5 directs basal cells towards a luminal cell fate, and thereby results in a marked decrease of basal MEC progenitor activity in vitro and in vivo. Mechanistically, these effects are mediated through β4-integrin and activation of p21 (encoded by CDKN1A). Thus, we demonstrate that laminin matrix adhesion is a key determinant of basal identity and essential to building and maintaining a functional multicellular epithelium.
Collapse
Affiliation(s)
- Johanna I. Englund
- Institute of Biotechnology, HiLIFE, University of Helsinki 1 , Helsinki FI-00014 , Finland
| | - Hien Bui
- Institute of Biotechnology, HiLIFE, University of Helsinki 1 , Helsinki FI-00014 , Finland
| | - Defne D. Dinç
- Institute of Biomedicine, Cancer Laboratory FICAN west, University of Turku 2 , Turku FI-20014 , Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University 3 , Turku FI-20014 , Finland
| | - Oona Paavolainen
- Institute of Biomedicine, Cancer Laboratory FICAN west, University of Turku 2 , Turku FI-20014 , Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University 3 , Turku FI-20014 , Finland
| | - Tomás McKenna
- Karolinska Institutet 4 Department of Cell and Molecular Biology (CMB) , , Stockholm SE-171 77 , Sweden
| | - Suvi Laitinen
- Institute of Biotechnology, HiLIFE, University of Helsinki 1 , Helsinki FI-00014 , Finland
| | - Pauliina Munne
- Finnish Cancer Institute, FICAN South Helsinki University Hospital & Translational Cancer Medicine, Medical Faculty, University of Helsinki 5 , Helsinki FI-00014 , Finland
| | - Juha Klefström
- Finnish Cancer Institute, FICAN South Helsinki University Hospital & Translational Cancer Medicine, Medical Faculty, University of Helsinki 5 , Helsinki FI-00014 , Finland
| | - Emilia Peuhu
- Institute of Biomedicine, Cancer Laboratory FICAN west, University of Turku 2 , Turku FI-20014 , Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University 3 , Turku FI-20014 , Finland
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki 1 , Helsinki FI-00014 , Finland
- Karolinska Institutet 4 Department of Cell and Molecular Biology (CMB) , , Stockholm SE-171 77 , Sweden
- University of Helsinki 6 Faculty of Biological and Environmental Sciences , , Helsinki FI-00014 , Finland
| |
Collapse
|
7
|
van Ineveld RL, van Vliet EJ, Wehrens EJ, Alieva M, Rios AC. 3D imaging for driving cancer discovery. EMBO J 2022; 41:e109675. [PMID: 35403737 PMCID: PMC9108604 DOI: 10.15252/embj.2021109675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 11/09/2022] Open
Abstract
Our understanding of the cellular composition and architecture of cancer has primarily advanced using 2D models and thin slice samples. This has granted spatial information on fundamental cancer biology and treatment response. However, tissues contain a variety of interconnected cells with different functional states and shapes, and this complex organization is impossible to capture in a single plane. Furthermore, tumours have been shown to be highly heterogenous, requiring large-scale spatial analysis to reliably profile their cellular and structural composition. Volumetric imaging permits the visualization of intact biological samples, thereby revealing the spatio-phenotypic and dynamic traits of cancer. This review focuses on new insights into cancer biology uniquely brought to light by 3D imaging and concomitant progress in cancer modelling and quantitative analysis. 3D imaging has the potential to generate broad knowledge advance from major mechanisms of tumour progression to new strategies for cancer treatment and patient diagnosis. We discuss the expected future contributions of the newest imaging trends towards these goals and the challenges faced for reaching their full application in cancer research.
Collapse
Affiliation(s)
- Ravian L van Ineveld
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Esmée J van Vliet
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Maria Alieva
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| |
Collapse
|
8
|
Sahu S, Albaugh ME, Martin BK, Patel NL, Riffle L, Mackem S, Kalen JD, Sharan SK. Growth factor dependency in mammary organoids regulates ductal morphogenesis during organ regeneration. Sci Rep 2022; 12:7200. [PMID: 35504930 PMCID: PMC9065107 DOI: 10.1038/s41598-022-11224-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/20/2022] [Indexed: 12/28/2022] Open
Abstract
Signaling pathways play an important role in cell fate determination in stem cells and regulate a plethora of developmental programs, the dysregulation of which can lead to human diseases. Growth factors (GFs) regulating these signaling pathways therefore play a major role in the plasticity of adult stem cells and modulate cellular differentiation and tissue repair outcomes. We consider murine mammary organoid generation from self-organizing adult stem cells as a tool to understand the role of GFs in organ development and tissue regeneration. The astounding capacity of mammary organoids to regenerate a gland in vivo after transplantation makes it a convenient model to study organ regeneration. We show organoids grown in suspension with minimal concentration of Matrigel and in the presence of a cocktail of GFs regulating EGF and FGF signaling can recapitulate key epithelial layers of adult mammary gland. We establish a toolkit utilizing in vivo whole animal imaging and ultrasound imaging combined with ex vivo approaches including tissue clearing and confocal imaging to study organ regeneration and ductal morphogenesis. Although the organoid structures were severely impaired in vitro when cultured in the presence of individual GFs, ex vivo imaging revealed ductal branching after transplantation albeit with significantly reduced number of terminal end buds. We anticipate these imaging modalities will open novel avenues to study mammary gland morphogenesis in vivo and can be beneficial for monitoring mammary tumor progression in pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program, Centre for Cancer Research, National Cancer Institute, Bldg- 560, Room 32-33, 1050 Boyles Street, Frederick, MD, 21702, USA
| | - Mary E Albaugh
- Mouse Cancer Genetics Program, Centre for Cancer Research, National Cancer Institute, Bldg- 560, Room 32-33, 1050 Boyles Street, Frederick, MD, 21702, USA
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Betty K Martin
- Mouse Cancer Genetics Program, Centre for Cancer Research, National Cancer Institute, Bldg- 560, Room 32-33, 1050 Boyles Street, Frederick, MD, 21702, USA
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Nimit L Patel
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Lisa Riffle
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Susan Mackem
- Cancer and Developmental Biology Laboratory, Centre for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Joseph D Kalen
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, Centre for Cancer Research, National Cancer Institute, Bldg- 560, Room 32-33, 1050 Boyles Street, Frederick, MD, 21702, USA.
- Centre for Advanced Preclinical Research, National Cancer Institute, Bldg- 560, Room 32-33, 1050 Boyles Street, Frederick, MD, 21702, USA.
| |
Collapse
|
9
|
Bartlett AP, Harman RM, Weiss JR, Van de Walle GR. Establishment and characterization of equine mammary organoids using a method translatable to other non-traditional model species. Development 2022; 149:274742. [DOI: 10.1242/dev.200412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/21/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Mammary organoid (MaO) models are only available for a few traditional model organisms, limiting our ability to investigate mammary gland development and cancer across mammals. This study established equine mammary organoids (EqMaOs) from cryopreserved mammary tissue, in which mammary tissue fragments were isolated and embedded into a 3D matrix to produce EqMaOs. We evaluated viability, proliferation and budding capacity of EqMaOs at different time points during culture, showing that although the number of proliferative cells decreased over time, viability was maintained and budding increased. We further characterized EqMaOs based on expression of stem cell, myoepithelial and luminal markers, and found that EqMaOs expressed these markers throughout culture and that a bilayered structure as seen in vivo was recapitulated. We used the milk-stimulating hormone prolactin to induce milk production, which was verified by the upregulation of milk proteins, most notably β-casein. Additionally, we showed that our method is also applicable to additional non-traditional mammalian species, particularly domesticated animals such as cats, pigs and rabbits. Collectively, MaO models across species will be a useful tool for comparative developmental and cancer studies.
Collapse
Affiliation(s)
- Arianna P. Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Rebecca M. Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Jennifer R. Weiss
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
10
|
Fei K, Zhang J, Yuan J, Xiao P. Present Application and Perspectives of Organoid Imaging Technology. Bioengineering (Basel) 2022; 9:121. [PMID: 35324810 PMCID: PMC8945799 DOI: 10.3390/bioengineering9030121] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/23/2022] [Accepted: 03/13/2022] [Indexed: 11/18/2022] Open
Abstract
An organoid is a miniaturized and simplified in vitro model with a similar structure and function to a real organ. In recent years, the use of organoids has increased explosively in the field of growth and development, disease simulation, drug screening, cell therapy, etc. In order to obtain necessary information, such as morphological structure, cell function and dynamic signals, it is necessary and important to directly monitor the culture process of organoids. Among different detection technologies, imaging technology is a simple and convenient choice and can realize direct observation and quantitative research. In this review, the principle, advantages and disadvantages of imaging technologies that have been applied in organoids research are introduced. We also offer an overview of prospective technologies for organoid imaging. This review aims to help biologists find appropriate imaging techniques for different areas of organoid research, and also contribute to the development of organoid imaging systems.
Collapse
Affiliation(s)
| | | | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| | - Peng Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| |
Collapse
|
11
|
Caruso M, Huang S, Mourao L, Scheele CLGJ. A Mammary Organoid Model to Study Branching Morphogenesis. Front Physiol 2022; 13:826107. [PMID: 35399282 PMCID: PMC8988230 DOI: 10.3389/fphys.2022.826107] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/31/2022] [Indexed: 11/17/2022] Open
Abstract
Branching morphogenesis is the process that gives rise to branched structures in several organs, such as the lung, the kidney, and the mammary gland. Although morphologically well described, the exact mechanisms driving branch elongation and bifurcation are still poorly understood. Signaling cues from the stroma and extracellular matrix have an important role in driving branching morphogenesis. Organoid models derived from primary mammary epithelial cells have emerged as a powerful tool to gain insight into branching morphogenesis of the mammary gland. However, current available mammary organoid culture protocols result in morphologically simple structures which do not resemble the complex branched structure of the in vivo mammary gland. Supplementation of growth factors to mammary organoids cultured in basement membrane extract or collagen I were shown to induce bud formation and elongation but are not sufficient to drive true branching events. Here, we present an improved culture approach based on 3D primary mammary epithelial cell culture to develop branched organoids with a complex morphology. By alternating the addition of fibroblast growth factor 2 and epidermal growth factor to mammary organoids cultured in a basement membrane extract matrix enriched with collagen type I fibers, we obtain complex mammary organoid structures with primary, secondary, and tertiary branches over a period of 15-20 days. Mammary organoid structures grow >1 mm in size and show an elongated and branched shape which resembles in vivo mammary gland morphology. This novel branched mammary organoid model offers many possibilities to study the mechanisms of branching in the developing mammary gland.
Collapse
Affiliation(s)
- Marika Caruso
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Sjanie Huang
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Larissa Mourao
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Colinda L. G. J. Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Brenna C, Simioni C, Varano G, Conti I, Costanzi E, Melloni M, Neri LM. Optical tissue clearing associated with 3D imaging: application in preclinical and clinical studies. Histochem Cell Biol 2022; 157:497-511. [PMID: 35235045 PMCID: PMC9114043 DOI: 10.1007/s00418-022-02081-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 12/23/2022]
Abstract
Understanding the inner morphology of intact tissues is one of the most competitive challenges in modern biology. Since the beginning of the twentieth century, optical tissue clearing (OTC) has provided solutions for volumetric imaging, allowing the microscopic visualization of thick sections of tissue, organoids, up to whole organs and organisms (for example, mouse or rat). Recently, tissue clearing has also been introduced in clinical settings to achieve a more accurate diagnosis with the support of 3D imaging. This review aims to give an overview of the most recent developments in OTC and 3D imaging and to illustrate their role in the field of medical diagnosis, with a specific focus on clinical applications.
Collapse
Affiliation(s)
- Cinzia Brenna
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy.,Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Carolina Simioni
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121, Ferrara, Italy.,LTTA - Electron Microscopy Center, University of Ferrara, 44121, Ferrara, Italy
| | - Gabriele Varano
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Ilaria Conti
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Eva Costanzi
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Luca Maria Neri
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy. .,LTTA - Electron Microscopy Center, University of Ferrara, 44121, Ferrara, Italy.
| |
Collapse
|
13
|
Heitink L, Whittle JR, Vaillant F, Capaldo BD, Dekkers JF, Dawson CA, Milevskiy MJG, Surgenor E, Tsai M, Chen H, Christie M, Chen Y, Smyth GK, Herold MJ, Strasser A, Lindeman GJ, Visvader JE. In vivo genome-editing screen identifies tumor suppressor genes that cooperate with Trp53 loss during mammary tumorigenesis. Mol Oncol 2022; 16:1119-1131. [PMID: 35000262 PMCID: PMC8895454 DOI: 10.1002/1878-0261.13179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/07/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022] Open
Abstract
Breast cancer is a heterogeneous disease that comprises multiple histological and molecular subtypes. To gain insight into mutations that drive breast tumorigenesis, we describe a pipeline for the identification and validation of tumor suppressor genes. Based on an in vivo genome‐wide CRISPR/Cas9 screen in Trp53+/– heterozygous mice, we identified tumor suppressor genes that included the scaffold protein Axin1, the protein kinase A regulatory subunit gene Prkar1a, as well as the proof‐of‐concept genes Pten, Nf1, and Trp53 itself. Ex vivo editing of primary mammary epithelial organoids was performed to further interrogate the roles of Axin1 and Prkar1a. Increased proliferation and profound changes in mammary organoid morphology were observed for Axin1/Trp53 and Prkar1a/Trp53 double mutants compared to Pten/Trp53 double mutants. Furthermore, direct in vivo genome editing via intraductal injection of lentiviruses engineered to express dual short‐guide RNAs revealed that mutagenesis of Trp53 and either Prkar1a, Axin1, or Pten markedly accelerated tumor development compared to Trp53‐only mutants. This proof‐of‐principle study highlights the application of in vivo CRISPR/Cas9 editing for uncovering cooperativity between defects in tumor suppressor genes that elicit mammary tumorigenesis.
Collapse
Affiliation(s)
- Luuk Heitink
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
| | - James R. Whittle
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneAustralia
| | - François Vaillant
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
| | - Bianca D. Capaldo
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
| | - Johanna F. Dekkers
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Caleb A. Dawson
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Immunology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Michael J. G. Milevskiy
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
| | - Elliot Surgenor
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Minhsuang Tsai
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Huei‐Rong Chen
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Michael Christie
- Personalised Oncology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of PathologyThe Royal Melbourne HospitalParkvilleAustralia
| | - Yunshun Chen
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Gordon K. Smyth
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- School of Mathematics and StatisticsThe University of MelbourneParkvilleAustralia
| | - Marco J. Herold
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Blood Cells and Blood Cancer DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Andreas Strasser
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Blood Cells and Blood Cancer DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Geoffrey J. Lindeman
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneAustralia
| | - Jane E. Visvader
- ACRF Cancer Biology and Stem Cells DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical BiologyThe University of MelbourneParkvilleAustralia
| |
Collapse
|
14
|
Lan Q, Satta J, Myllymäki SM, Trela E, Lindström R, Kaczyńska B, Englund J, Mikkola ML. Protocol for Studying Embryonic Mammary Gland Branching Morphogenesis Ex Vivo. Methods Mol Biol 2022; 2471:1-18. [PMID: 35175589 DOI: 10.1007/978-1-0716-2193-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mammary gland development starts during embryogenesis, and the process continues after birth. During development, the mammary gland undergoes massive morphological and physiological alterations including growth, invasion, and branching morphogenesis providing an ideal model for stem cell and cancer biology studies. Great efforts have been made in understanding mammary gland development during puberty and adulthood; however, the process during embryogenesis is still elusive. One reason is that the tools to study tissue dynamics during development are limited, which is partially due to the lack of an ex vivo culture method. Here we describe an updated organ culture protocol of the murine embryonic mammary gland. This powerful tool allows monitoring of growth and branching morphogenesis of mammary gland ex vivo by live imaging. In addition, we introduce a novel method for culturing intact, stroma-free mammary rudiments from late gestation mouse embryos in 3D in Matrigel. This approach can be used to identify the direct stromal cues for branching morphogenesis.
Collapse
Affiliation(s)
- Qiang Lan
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jyoti Satta
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Satu-Marja Myllymäki
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Ewelina Trela
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Riitta Lindström
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Beata Kaczyńska
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Johanna Englund
- Centre of Excellence in Stem Cell Metabolism, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Marja L Mikkola
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
15
|
El Khoury R, Nagiah N, Mudloff JA, Thakur V, Chattopadhyay M, Joddar B. 3D Bioprinted Spheroidal Droplets for Engineering the Heterocellular Coupling between Cardiomyocytes and Cardiac Fibroblasts. CYBORG AND BIONIC SYSTEMS 2021; 2021:9864212. [PMID: 35795473 PMCID: PMC9254634 DOI: 10.34133/2021/9864212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Since conventional human cardiac two-dimensional (2D) cell culture and multilayered three-dimensional (3D) models fail in recapitulating cellular complexity and possess inferior translational capacity, we designed and developed a high-throughput scalable 3D bioprinted cardiac spheroidal droplet-organoid model with cardiomyocytes and cardiac fibroblasts that can be used for drug screening or regenerative engineering applications. This study helped establish the parameters for bioprinting and cross-linking a gelatin-alginate-based bioink into 3D spheroidal droplets. A flattened disk-like structure developed in prior studies from our laboratory was used as a control. The microstructural and mechanical stability of the 3D spheroidal droplets was assessed and was found to be ideal for a cardiac scaffold. Adult human cardiac fibroblasts and AC16 cardiomyocytes were mixed in the bioink and bioprinted. Live-dead assay and flow cytometry analysis revealed robust biocompatibility of the 3D spheroidal droplets that supported the growth and proliferation of the cardiac cells in the long-term cultures. Moreover, the heterocellular gap junctional coupling between the cardiomyocytes and cardiac fibroblasts further validated the 3D cardiac spheroidal droplet model.
Collapse
Affiliation(s)
- Raven El Khoury
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Naveen Nagiah
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Joel A. Mudloff
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | - Vikram Thakur
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Munmun Chattopadhyay
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| |
Collapse
|
16
|
Mohan SC, Lee TY, Giuliano AE, Cui X. Current Status of Breast Organoid Models. Front Bioeng Biotechnol 2021; 9:745943. [PMID: 34805107 PMCID: PMC8602090 DOI: 10.3389/fbioe.2021.745943] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/22/2021] [Indexed: 11/28/2022] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women globally. Although mouse models have been critical in advancing the knowledge of BC tumorigenesis and progression, human breast models comprising the breast tissue microenvironment are needed to help elucidate the underlying mechanisms of BC risk factors. As such, it is essential to identify an ex vivo human breast tissue mimetic model that can accurately pinpoint the effects of these factors in BC development. While two-dimensional models have been invaluable, they are not suitable for studying patient-specific tumor biology and drug response. Recent developments in three-dimensional (3D) models have led to the prominence of organized structures grown in a 3D environment called “organoids.” Breast organoids can accurately recapitulate the in vivo breast microenvironment and have been used to examine factors that affect signaling transduction, gene expression, and tissue remodeling. In this review, the applications, components, and protocols for development of breast organoids are discussed. We summarize studies that describe the utility of breast organoids, including in the study of normal mammary gland development and tumorigenesis. Finally, we provide an overview of protocols for development of breast organoids, and the advantages and disadvantages of different techniques in studies are described. The included studies have shown that breast organoids will continue to serve as a crucial platform for understanding of progression of BC tumors and the testing of novel therapeutics.
Collapse
Affiliation(s)
- Srivarshini Cherukupalli Mohan
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Tian-Yu Lee
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Armando E Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
17
|
Fang G, Lu H, Rodriguez de la Fuente L, Law AMK, Lin G, Jin D, Gallego‐Ortega D. Mammary Tumor Organoid Culture in Non-Adhesive Alginate for Luminal Mechanics and High-Throughput Drug Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102418. [PMID: 34494727 PMCID: PMC8564453 DOI: 10.1002/advs.202102418] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/25/2021] [Indexed: 05/14/2023]
Abstract
Mammary tumor organoids have become a promising in vitro model for drug screening and personalized medicine. However, the dependency on the basement membrane extract (BME) as the growth matrices limits their comprehensive application. In this work, mouse mammary tumor organoids are established by encapsulating tumor pieces in non-adhesive alginate. High-throughput generation of organoids in alginate microbeads is achieved utilizing microfluidic droplet technology. Tumor pieces within the alginate microbeads developed both luminal- and solid-like structures and displayed a high similarity to the original fresh tumor in cellular phenotypes and lineages. The mechanical forces of the luminal organoids in the alginate capsules are analyzed with the theory of the thick-wall pressure vessel (TWPV) model. The luminal pressure of the organoids increase with the lumen growth and can reach 2 kPa after two weeks' culture. Finally, the mammary tumor organoids are treated with doxorubicin and latrunculin A to evaluate their application as a drug screening platform. It is found that the drug response is related to the luminal size and pressures of organoids. This high-throughput culture for mammary tumor organoids may present a promising tool for preclinical drug target validation and personalized medicine.
Collapse
Affiliation(s)
- Guocheng Fang
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Hongxu Lu
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Laura Rodriguez de la Fuente
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
| | - Andrew M. K. Law
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
| | - Gungun Lin
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Dayong Jin
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
- UTS‐SUSTech Joint Research Centre for Biomedical Materials and DevicesDepartment of Biomedical EngineeringSouthern University of Science and TechnologyShenzhenGuangdong518055China
| | - David Gallego‐Ortega
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
- School of Biomedical EngineeringFaculty of EngineeringUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| |
Collapse
|
18
|
Abstract
3D cultures of mammary epithelial cells purified from murine models provide a unique resource to study genetically defined breast cancer and response to targeted therapies. Here, we describe step-by-step experimental procedures for the successful establishment of murine mammary organoid lines isolated from mammary glands or mammary tumors driven by mutations in components of the PI3K pathway. These detailed protocols also include procedures to perform assays that can be adopted to screen response to drug treatments and to inform better therapies. For details on potential applications and use of this protocol, please refer to Yip et al. (2020).
Collapse
Affiliation(s)
- Hon Yan Kelvin Yip
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
19
|
Wrenn E, Huang Y, Cheung K. Collective metastasis: coordinating the multicellular voyage. Clin Exp Metastasis 2021; 38:373-399. [PMID: 34254215 PMCID: PMC8346286 DOI: 10.1007/s10585-021-10111-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
The metastatic process is arduous. Cancer cells must escape the confines of the primary tumor, make their way into and travel through the circulation, then survive and proliferate in unfavorable microenvironments. A key question is how cancer cells overcome these multiple barriers to orchestrate distant organ colonization. Accumulating evidence in human patients and animal models supports the hypothesis that clusters of tumor cells can complete the entire metastatic journey in a process referred to as collective metastasis. Here we highlight recent studies unraveling how multicellular coordination, via both physical and biochemical coupling of cells, induces cooperative properties advantageous for the completion of metastasis. We discuss conceptual challenges and unique mechanisms arising from collective dissemination that are distinct from single cell-based metastasis. Finally, we consider how the dissection of molecular transitions regulating collective metastasis could offer potential insight into cancer therapy.
Collapse
Affiliation(s)
- Emma Wrenn
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, 98195, USA
| | - Yin Huang
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Kevin Cheung
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
20
|
Slepicka PF, Somasundara AVH, Dos Santos CO. The molecular basis of mammary gland development and epithelial differentiation. Semin Cell Dev Biol 2021; 114:93-112. [PMID: 33082117 PMCID: PMC8052380 DOI: 10.1016/j.semcdb.2020.09.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
21
|
Gritti N, Oriola D, Trivedi V. Rethinking embryology in vitro: A synergy between engineering, data science and theory. Dev Biol 2021; 474:48-61. [DOI: 10.1016/j.ydbio.2020.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
|
22
|
Charifou E, Sumbal J, Koledova Z, Li H, Chiche A. A Robust Mammary Organoid System to Model Lactation and Involution-like Processes. Bio Protoc 2021; 11:e3996. [PMID: 34124297 PMCID: PMC8160540 DOI: 10.21769/bioprotoc.3996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 11/02/2022] Open
Abstract
The mammary gland is a highly dynamic tissue that changes throughout reproductive life, including growth during puberty and repetitive cycles of pregnancy and involution. Mammary gland tumors represent the most common cancer diagnosed in women worldwide. Studying the regulatory mechanisms of mammary gland development is essential for understanding how dysregulation can lead to breast cancer initiation and progression. Three-dimensional (3D) mammary organoids offer many exciting possibilities for the study of tissue development and breast cancer. In the present protocol derived from Sumbal et al., we describe a straightforward 3D organoid system for the study of lactation and involution ex vivo. We use primary and passaged mouse mammary organoids stimulated with fibroblast growth factor 2 (FGF2) and prolactin to model the three cycles of mouse mammary gland lactation and involution processes. This 3D organoid model represents a valuable tool to study late postnatal mammary gland development and breast cancer, in particular postpartum-associated breast cancer. Graphic abstract: Mammary gland organoid isolation and culture procedures.
Collapse
Affiliation(s)
- Elsa Charifou
- Cellular Plasticity & Disease Modeling - Department of Developmental & Stem Cell Biology, CNRS UMR3738 - Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| | - Jakub Sumbal
- Cellular Plasticity & Disease Modeling - Department of Developmental & Stem Cell Biology, CNRS UMR3738 - Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno 625 00, Czech Republic
| | - Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno 625 00, Czech Republic
| | - Han Li
- Cellular Plasticity & Disease Modeling - Department of Developmental & Stem Cell Biology, CNRS UMR3738 - Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| | - Aurélie Chiche
- Cellular Plasticity & Disease Modeling - Department of Developmental & Stem Cell Biology, CNRS UMR3738 - Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| |
Collapse
|
23
|
Junyent S, Reeves J, Gentleman E, Habib SJ. Pluripotency state regulates cytoneme selectivity and self-organization of embryonic stem cells. J Cell Biol 2021; 220:e202005095. [PMID: 33606876 PMCID: PMC7903188 DOI: 10.1083/jcb.202005095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/09/2020] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
To coordinate cell fate with changes in spatial organization, stem cells (SCs) require specific and adaptable systems of signal exchange and cell-to-cell communication. Pluripotent embryonic stem cells (ESCs) use cytonemes to pair with trophoblast stem cells (TSCs) and form synthetic embryonic structures in a Wnt-dependent manner. How these interactions vary with pluripotency states remains elusive. Here we show that ESC transition to an early primed ESC (pESC) state reduces their pairing with TSCs and impairs synthetic embryogenesis. pESCs can activate the Wnt/β-catenin pathway in response to soluble Wnt ligands, but their cytonemes form unspecific and unstable interactions with localized Wnt sources. This is due to an impaired crosstalk between Wnt and glutamate receptor activity and reduced generation of Ca2+ transients on the cytonemes upon Wnt source contact. Induced iGluR activation can partially restore cytoneme function in pESCs, while transient overexpression of E-cadherin improves pESC-TSC pairing. Our results illustrate how changes in pluripotency state alter the mechanisms SCs use to self-organize.
Collapse
Affiliation(s)
- Sergi Junyent
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Joshua Reeves
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Shukry J. Habib
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| |
Collapse
|
24
|
Dawson CA, Mueller SN, Lindeman GJ, Rios AC, Visvader JE. Intravital microscopy of dynamic single-cell behavior in mouse mammary tissue. Nat Protoc 2021; 16:1907-1935. [PMID: 33627843 DOI: 10.1038/s41596-020-00473-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/24/2020] [Indexed: 01/31/2023]
Abstract
Multiphoton intravital imaging is essential for understanding cellular behavior and function in vivo. The adipose-rich environment of the mammary gland poses a unique challenge to in vivo microscopy due to light scattering that impedes high-resolution imaging. Here we provide a protocol for high-quality, six-color 3D intravital imaging of regions across the entire mouse mammary gland and associated tissues for several hours while maintaining tissue access for microdissection and labeling. An incision at the ventral midline and along the right hind leg creates a skin flap that is then secured to a raised platform skin side down. This allows for fluorescence-guided microdissection of connective tissue to provide unimpeded imaging of mammary ducts. A sealed imaging chamber over the skin flap creates a stable environment while maintaining access to large tissue regions for imaging with an upright microscope. We provide a strategy for imaging single cells and the tissue microenvironment utilizing multicolor Confetti lineage-tracing and additional dyes using custom-designed filters and sequential excitation with dual multiphoton lasers. Furthermore, we describe a strategy for simultaneous imaging and photomanipulation of single cells using the Olympus SIM scanner and provide steps for 3D video processing, visualization and high-dimensional analysis of single-cell behavior. We then provide steps for multiplexing intravital imaging with fixation, immunostaining, tissue clearing and 3D confocal imaging to associate cell behavior with protein expression. The skin-flap surgery and chamber preparation take 1.5 h, followed by up to 12 h of imaging. Applications range from basic filming in 1 d to 5 d for multiplexing and complex analysis.
Collapse
Affiliation(s)
- Caleb A Dawson
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Melbourne, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Anne C Rios
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
25
|
Wang J, Song W, Yang R, Li C, Wu T, Dong XB, Zhou B, Guo X, Chen J, Liu Z, Yu QC, Li W, Fu J, Zeng YA. Endothelial Wnts control mammary epithelial patterning via fibroblast signaling. Cell Rep 2021; 34:108897. [PMID: 33789106 DOI: 10.1016/j.celrep.2021.108897] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/09/2020] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Endothelial and fibroblast niches are crucial for epithelial organs. How these heterotypic cells interact is of great interest. In this study, we reveal an axis of signaling in which fibroblasts relay Wnt signals from the endothelial niche to organize epithelial patterning. We generate an Axin2-membrane GFP (mGFP) reporter mouse and observe robust Wnt/β-catenin signaling activities in fibroblasts surrounding the mammary epithelium. To enable cell-type-specific gene manipulation in vitro, we establish an organoid system via coculture of endothelial cells (ECs), fibroblasts, and mammary epithelial cells. Deletion of β-catenin in fibroblasts impedes epithelium branching, and ECs are responsible for the activation of Wnt/β-catenin signaling in fibroblasts. In vivo, EC deletion of Wntless inhibits Wnt/β-catenin signaling activity in fibroblasts, rendering a reduction in epithelial branches. These findings highlight the significance of the endothelial niche in tissue patterning, shedding light on the interactive mechanisms in which distinct niche components orchestrate epithelial organogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Jingqiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Wenqian Song
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Ruikai Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiao Bing Dong
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xizhi Guo
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Wen Li
- Center of reproductive medicine, Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Junfen Fu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China.
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
26
|
Zahmatkesh E, Khoshdel-Rad N, Mirzaei H, Shpichka A, Timashev P, Mahmoudi T, Vosough M. Evolution of organoid technology: Lessons learnt in Co-Culture systems from developmental biology. Dev Biol 2021; 475:37-53. [PMID: 33684433 DOI: 10.1016/j.ydbio.2021.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
In recent years, the development of 3D organoids has opened new avenues of investigation into development, physiology, and regenerative medicine. Organoid formation and the process of organogenesis share common developmental pathways; thus, our knowledge of developmental biology can help model the complexity of different organs to refine organoids into a more sophisticated platform. The developmental process is strongly dependent on complex networks and communication of cell-cell and cell-matrix interactions among different cell populations and their microenvironment, during embryogenesis. These interactions affect cell behaviors such as proliferation, survival, migration, and differentiation. Co-culture systems within the organoid technology were recently developed and provided the highly physiologically relevant systems. Supportive cells including various types of endothelial and stromal cells provide the proper microenvironment, facilitate organoid assembly, and improve vascularization and maturation of organoids. This review discusses the role of the co-culture systems in organoid generation, with a focus on how knowledge of developmental biology has directed and continues to shape the development of more evolved 3D co-culture system-derived organoids.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia; Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, N.N.Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
27
|
Dekkers JF, Whittle JR, Vaillant F, Chen HR, Dawson C, Liu K, Geurts MH, Herold MJ, Clevers H, Lindeman GJ, Visvader JE. Modeling Breast Cancer Using CRISPR-Cas9-Mediated Engineering of Human Breast Organoids. J Natl Cancer Inst 2021; 112:540-544. [PMID: 31589320 DOI: 10.1093/jnci/djz196] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/04/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is characterized by histological and functional heterogeneity, posing a clinical challenge for patient treatment. Emerging evidence suggests that the distinct subtypes reflect the repertoire of genetic alterations and the target cell. However, the precise initiating events that predispose normal epithelium to neoplasia are poorly understood. Here, we demonstrate that breast epithelial organoids can be generated from human reduction mammoplasties (12 out of 12 donors), thus creating a tool to study the clonal evolution of breast cancer. To recapitulate de novo oncogenesis, we exploited clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 for targeted knockout of four breast cancer-associated tumor suppressor genes (P53, PTEN, RB1, NF1) in mammary progenitor cells from six donors. Mutant organoids gained long-term culturing capacity and formed estrogen-receptor positive luminal tumors on transplantation into mice for one out of six P53/PTEN/RB1-mutated and three out of six P53/PTEN/RB1/NF1-mutated lines. These organoids responded to endocrine therapy or chemotherapy, supporting the potential utility of this model to enhance our understanding of the molecular events that culminate in specific subtypes of breast cancer.
Collapse
Affiliation(s)
- Johanna F Dekkers
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - James R Whittle
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology.,Department of Medicine (GHL), The University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - François Vaillant
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - Huei-Rong Chen
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - Caleb Dawson
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - Kevin Liu
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - Maarten H Geurts
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands
| | - Marco J Herold
- Blood Cells and Blood Cancer Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center, Utrecht, the Netherlands
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division.,Department of Medicine (GHL), The University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology
| |
Collapse
|
28
|
Gu VW, Cho E, Thompson DT, Cassady VC, Borcherding N, Koch KE, Wu VT, Lorenzen AW, van der Heide DM, White JR, Kulak MV, Williams T, Zhang W, Weigel RJ. AP-2γ Is Required for Maintenance of Multipotent Mammary Stem Cells. Stem Cell Reports 2020; 16:106-119. [PMID: 33382976 PMCID: PMC7897584 DOI: 10.1016/j.stemcr.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/13/2023] Open
Abstract
Mammary gland ductal morphogenesis depends on the differentiation of mammary stem cells (MaSCs) into basal and luminal lineages. The AP-2γ transcription factor, encoded by Tfap2c, has a central role in mammary gland development but its effect in mammary lineages and specifically MaSCs is largely unknown. Here, we utilized an inducible, conditional knockout of Tfap2c to elucidate the role of AP-2γ in maintenance and differentiation of MaSCs. Loss of AP-2γ in the basal epithelium profoundly altered the transcriptomes and decreased the number of cells within several clusters of mammary epithelial cells, including adult MaSCs and luminal progenitors. AP-2γ regulated the expression of genes known to be required for mammary development, including Cebpb, Nfkbia, and Rspo1. As a result, AP-2γ-deficient mice exhibited repressed mammary gland ductal outgrowth and inhibition of regenerative capacity. The findings demonstrate that AP-2γ can regulate development of mammary gland structures potentially regulating maintenance and differentiation of multipotent MaSCs. AP-2γ-deficient mice exhibit repressed ductal outgrowth and regenerative capacity Loss of AP-2γ reduced cells within mammary stem and luminal progenitor clusters AP-2γ target genes, including Cebpb, Nfkbia, and Rspo1, regulate mammary development AP-2γ potentially regulates maintenance of multipotent mammary stem cells
Collapse
Affiliation(s)
- Vivian W Gu
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Edward Cho
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Dakota T Thompson
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Victoria C Cassady
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | | | - Kelsey E Koch
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Vincent T Wu
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Allison W Lorenzen
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Dana M van der Heide
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Jeffrey R White
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Mikhail V Kulak
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Ronald J Weigel
- Department of Surgery, University of Iowa, 200 Hawkins Drive, JCP 1509 Iowa City, IA 52242-1086, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
29
|
Sumbal J, Budkova Z, Traustadóttir GÁ, Koledova Z. Mammary Organoids and 3D Cell Cultures: Old Dogs with New Tricks. J Mammary Gland Biol Neoplasia 2020; 25:273-288. [PMID: 33210256 DOI: 10.1007/s10911-020-09468-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022] Open
Abstract
3D cell culture methods have been an integral part of and an essential tool for mammary gland and breast cancer research for half a century. In fact, mammary gland researchers, who discovered and deciphered the instructive role of extracellular matrix (ECM) in mammary epithelial cell functional differentiation and morphogenesis, were the pioneers of the 3D cell culture techniques, including organoid cultures. The last decade has brought a tremendous increase in the 3D cell culture techniques, including modifications and innovations of the existing techniques, novel biomaterials and matrices, new technological approaches, and increase in 3D culture complexity, accompanied by several redefinitions of the terms "3D cell culture" and "organoid". In this review, we provide an overview of the 3D cell culture and organoid techniques used in mammary gland biology and breast cancer research. We discuss their advantages, shortcomings and current challenges, highlight the recent progress in reconstructing the complex mammary gland microenvironment in vitro and ex vivo, and identify the missing 3D cell cultures, urgently needed to aid our understanding of mammary gland development, function, physiology, and disease, including breast cancer.
Collapse
Affiliation(s)
- Jakub Sumbal
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Budkova
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Gunnhildur Ásta Traustadóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland.
| | - Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
30
|
Wrenn ED, Moore BM, Greenwood E, McBirney M, Cheung KJ. Optimal, Large-Scale Propagation of Mouse Mammary Tumor Organoids. J Mammary Gland Biol Neoplasia 2020; 25:337-350. [PMID: 33106923 PMCID: PMC7587543 DOI: 10.1007/s10911-020-09464-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor organoids mimic the architecture and heterogeneity of in vivo tumors and enable studies of collective interactions between tumor cells as well as with their surrounding microenvironment. Although tumor organoids hold significant promise as cancer models, they are also more costly and labor-intensive to cultivate than traditional 2D cell culture. We sought to identify critical factors regulating organoid growth ex vivo, and to use these observations to develop a more efficient organoid expansion method. Using time-lapse imaging of mouse mammary tumor organoids in 3D culture, we observed that outgrowth potential varies non-linearly with initial organoid size. Maximal outgrowth occurred in organoids with a starting size between ~10 to 1000 cells. Based on these observations, we developed a suspension culture method that maintains organoids in the ideal size range, enabling expansion from 1 million to over 100 million cells in less than 2 weeks and less than 3 hours of hands-on time. Our method facilitates the rapid, cost-effective expansion of organoids for CRISPR based studies and other assays requiring a large amount of organoid starting material.
Collapse
Affiliation(s)
- Emma D Wrenn
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, 98195, USA
| | - Breanna M Moore
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Erin Greenwood
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Margaux McBirney
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Kevin J Cheung
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
31
|
Sahu S, Sharan SK. Translating Embryogenesis to Generate Organoids: Novel Approaches to Personalized Medicine. iScience 2020; 23:101485. [PMID: 32864586 PMCID: PMC7441954 DOI: 10.1016/j.isci.2020.101485] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The astounding capacity of pluripotent stem cells (PSCs) to differentiate and self-organize has revolutionized the development of 3D cell culture models. The major advantage is its ability to mimic in vivo microenvironments and cellular interactions when compared with the classical 2D cell culture models. Recent innovations in generating embryo-like structures (including blastoids and gastruloids) from PSCs have advanced the experimental accessibility to understand embryogenesis with immense potential to model human development. Taking cues on how embryonic development leads to organogenesis, PSCs can also be directly differentiated to form mini-organs or organoids of a particular lineage. Organoids have opened new avenues to augment our understanding of stem cell and regenerative biology, tissue homeostasis, and disease mechanisms. In this review, we provide insights from developmental biology with a comprehensive resource of signaling pathways that in a coordinated manner form embryo-like structures and organoids. Moreover, the advent of assembloids and multilineage organoids from PSCs opens a new dimension to study paracrine function and multi-tissue interactions in vitro. Although this led to an avalanche of enthusiasm to utilize organoids for organ transplantation studies, we examine the current limitations and provide perspectives to improve reproducibility, scalability, functional complexity, and cell-type characterization. Taken together, these 3D in vitro organ-specific and patient-specific models hold great promise for drug discovery, clinical management, and personalized medicine.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-04, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Shyam K. Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-33, 1050 Boyles Street, Frederick, MD 21702, USA
| |
Collapse
|
32
|
Zhang Y, Tang C, Span PN, Rowan AE, Aalders TW, Schalken JA, Adema GJ, Kouwer PHJ, Zegers MMP, Ansems M. Polyisocyanide Hydrogels as a Tunable Platform for Mammary Gland Organoid Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001797. [PMID: 32999851 PMCID: PMC7509700 DOI: 10.1002/advs.202001797] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Indexed: 05/20/2023]
Abstract
In the last decade, organoid technology has developed as a primary research tool in basic biological and clinical research. The reliance on poorly defined animal-derived extracellular matrix, however, severely limits its application in regenerative and translational medicine. Here, a well-defined, synthetic biomimetic matrix based on polyisocyanide (PIC) hydrogels that support efficient and reproducible formation of mammary gland organoids (MGOs) in vitro is presented. Only decorated with the adhesive peptide RGD for cell binding, PIC hydrogels allow MGO formation from mammary fragments or from purified single mammary epithelial cells. The cystic organoids maintain their capacity to branch for over two months, which is a fundamental and complex feature during mammary gland development. It is found that small variations in the 3D matrix give rise to large changes in the MGO: the ratio of the main cell types in the MGO is controlled by the cell-gel interactions via the cell binding peptide density, whereas gel stiffness controls colony formation efficiency, which is indicative of the progenitor density. Simple hydrogel modifications will allow for future introduction and customization of new biophysical and biochemical parameters, making the PIC platform an ideal matrix for in depth studies into organ development and for application in disease models.
Collapse
Affiliation(s)
- Ying Zhang
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 135NijmegenAJ 6525The Netherlands
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Chunling Tang
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Paul N. Span
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Alan E. Rowan
- Australian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLD4072Australia
| | - Tilly W. Aalders
- Experimental UrologyRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Jack A. Schalken
- Experimental UrologyRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Paul H. J. Kouwer
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 135NijmegenAJ 6525The Netherlands
| | - Mirjam M. P. Zegers
- Department of Cell BiologyRadboud Institute for Molecular SciencesRadboud University Medical CenterGeert Grooteplein 28NijmegenGA6525The Netherlands
| | - Marleen Ansems
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| |
Collapse
|
33
|
Hacker BC, Rafat M. Organoids as Complex In Vitro Models for Studying Radiation-Induced Cell Recruitment. Cell Mol Bioeng 2020; 13:341-357. [PMID: 32952734 PMCID: PMC7479086 DOI: 10.1007/s12195-020-00625-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023] Open
Abstract
Patients with triple negative breast cancer (TNBC) typically receive chemotherapy, surgery, and radiation therapy. Although this treatment improves prognosis for most patients, some patients continue to experience recurrence within 5 years. Preclinical studies have shown that immune cell infiltration at the irradiated site may play a significant role in tumor cell recruitment; however, little is known about the mechanisms that govern this process. This lack of knowledge highlights the need to evaluate radiation-induced cell infiltration with models that have controllable variables and maintain biological integrity. Mammary organoids are multicellular three-dimensional (3D) in vitro models, and they have been used to examine many aspects of mammary development and tumorigenesis. Organoids are also emerging as a powerful tool to investigate normal tissue radiation damage. In this review, we evaluate recent advances in mammary organoid technology, consider the advantages of using organoids to study radiation response, and discuss future directions for the applications of this technique.
Collapse
Affiliation(s)
- Benjamin C. Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
34
|
Tang C, van den Bijgaart RJE, Looman MWG, Triantis V, Nørskov Søndergaard J, Ansems M, Adema GJ. DC-SCRIPT affects mammary organoids branching morphogenesis by modulating the FGFR1-pERK signaling axis. Dev Biol 2020; 463:101-109. [PMID: 32422143 DOI: 10.1016/j.ydbio.2020.05.001] [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: 11/17/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 10/24/2022]
Abstract
Loss of expression of the transcription regulator DC-SCRIPT (Zfp366) is a prominent prognostic event in estrogen receptor-positive breast cancer patients. Studying the inherent link between breast morphogenesis and tumorigenesis, we recently reported that DC-SCRIPT affects normal mammary branching morphogenesis and mammary epithelium homeostasis. Here we investigated the molecular mechanism involved in DC-SCRIPT mediated regulation of FGF2 induced mammary branching morphogenesis in a 3D organoid culture system. Our data show that the delayed mammary organoid branching observed in DC-SCRIPT-/- organoids cannot be compensated for by increasing FGF2 levels. Interestingly, FGFR1, the dominant FGF2 receptor, was expressed at a significantly lower level in basal epithelial cells of DC-SCRIPT deficient organoids relative to wildtype organoids. A potential link between DC-SCRIPT and FGFR1 was further supported by the predicted locations of the DC-SCRIPT DNA binding motif at the Fgfr1 gene. Moreover, ERK1/2 phosphorylation downstream of the FGFR1 pathway was decreased in basal epithelial cells of DC-SCRIPT deficient organoids. Altogether, this study shows a relationship between DC-SCRIPT and FGFR1 related pERK signaling in modulating the branching morphogenesis of mammary organoids in vitro.
Collapse
Affiliation(s)
- Chunling Tang
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| | - Renske J E van den Bijgaart
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| | - Maaike W G Looman
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| | - Vassilis Triantis
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| | - Jonas Nørskov Søndergaard
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands; Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, 17177, Stockholm, Sweden.
| | - Marleen Ansems
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| | - Gosse J Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands.
| |
Collapse
|
35
|
Kudo KI, Takabatake M, Nagata K, Nishimura Y, Daino K, Iizuka D, Nishimura M, Suzuki K, Kakinuma S, Imaoka T. Flow Cytometry Definition of Rat Mammary Epithelial Cell Populations and Their Distinct Radiation Responses. Radiat Res 2020; 194:22-37. [PMID: 32352870 DOI: 10.1667/rr15566.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/08/2020] [Indexed: 11/03/2022]
Abstract
Breast tissue is very susceptible to radiation-induced carcinogenesis, and mammary stem/progenitor cells are potentially important targets of this. The mammary epithelium is maintained as two mostly independent lineages of luminal and basal cells. To elucidate their immediate radiation responses, we analyzed the mammary glands of female Sprague-Dawley rats, a radiation carcinogenesis model, using colony formation, flow cytometry and immunofluorescence. The results revealed that flow cytometry successfully fractionates rat mammary cells into CD49fhi CD24lo basal, CD49fmed CD24hi luminal progenitor, and CD49flo CD24hi mature luminal populations, resembling human breast, rather than mouse tissues. The colony-forming ability of the basal cells was more radiosensitive than the luminal progenitor cells. Flow cytometry and immunofluorescence showed more efficient cell cycle arrest, γ-H2AX responses, and apoptosis in the irradiated luminal progenitor cells, than in the basal cells. These results provide important insights into the early phase of radiation-induced breast cancer.
Collapse
Affiliation(s)
- Ken-Ichi Kudo
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masaru Takabatake
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kento Nagata
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yukiko Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kazuhiro Daino
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daisuke Iizuka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Keiji Suzuki
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
36
|
Corrò C, Novellasdemunt L, Li VSW. A brief history of organoids. Am J Physiol Cell Physiol 2020; 319:C151-C165. [PMID: 32459504 PMCID: PMC7468890 DOI: 10.1152/ajpcell.00120.2020] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/12/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
In vitro cell cultures are crucial research tools for modeling human development and diseases. Although the conventional monolayer cell cultures have been widely used in the past, the lack of tissue architecture and complexity of such model fails to inform the true biological processes in vivo. Recent advances in the organoid technology have revolutionized the in vitro culture tools for biomedical research by creating powerful three-dimensional (3D) models to recapitulate the cellular heterogeneity, structure, and functions of the primary tissues. Such organoid technology enables researchers to recreate human organs and diseases in a dish and thus holds great promises for many translational applications such as regenerative medicine, drug discovery, and precision medicine. In this review, we provide an overview of the organoid history and development. We discuss the strengths and limitations of organoids as well as their potential applications in the laboratory and the clinic.
Collapse
Affiliation(s)
- Claudia Corrò
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| | - Laura Novellasdemunt
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| |
Collapse
|
37
|
Organoid models for mammary gland dynamics and breast cancer. Curr Opin Cell Biol 2020; 66:51-58. [PMID: 32535255 DOI: 10.1016/j.ceb.2020.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/13/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
The mammary gland is a highly dynamic tissue that undergoes repeated cycles of growth and involution during pregnancy and menstruation. It is also the site from which breast cancers emerge. Organoids provide an in vitro model that preserves several of the cellular, structural, and microenvironmental features that dictate mammary gland function in vivo and have greatly advanced our understanding of glandular biology. Their tractability for genetic manipulation, live imaging, and high throughput screening have facilitated investigation into the mechanisms of glandular morphogenesis, structural maintenance, tumor progression, and invasion. Opportunities remain to enhance cellular and structural complexity of mammary organoid models, including incorporating additional cell types and hormone signaling.
Collapse
|
38
|
"Tissues in a Dish": A Review of Organoids in Plastic Surgery. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e2787. [PMID: 32440447 PMCID: PMC7209840 DOI: 10.1097/gox.0000000000002787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/25/2022]
Abstract
Organoids are in vitro miniaturized organ models—or, colloquially, “organs in a dish.” These 3-dimensional, multicellular structures are classically derived from pluripotent or multipotent stem cells. When guided by tissue-specific molecular factors, these cells exhibit self-organizing abilities that allow them to accurately recapitulate the architecture and function of the organ of interest. Organoid technology is a rapidly expanding field that endows researchers with an unprecedented ability to recreate, study, and manipulate complex biologic processes in vitro. When compared with standard 2- and 3-dimensional culture systems, which rely on co-culturing pre-established cell types, organoids provide a more biomimetic model with which to study the intercellular interactions necessary for in vivo organ function and architecture. Organoids have the potential to impact all avenues of medicine, including those fields most relevant to plastic and reconstructive surgery such as wound healing, oncology, craniofacial reconstruction, and burn care. In addition to their ability to serve as a novel tool for studying human-specific disease, organoids may be used for tissue engineering with the goal of developing biomimetic soft-tissue substitutes, which would be especially valuable to the plastic surgeon. Although organoids hold great promise for the field of plastic surgery, technical challenges in creating vascularized, multilineage organoids must be overcome to allow for the integration of this technology in clinical practice. This review provides a brief history of the organoid, highlights its potential clinical applications, discusses certain limitations, and examines the impact that this technology may have on the field of plastic and reconstructive surgery.
Collapse
|
39
|
Pereira EJ, Burns JS, Lee CY, Marohl T, Calderon D, Wang L, Atkins KA, Wang CC, Janes KA. Sporadic activation of an oxidative stress-dependent NRF2-p53 signaling network in breast epithelial spheroids and premalignancies. Sci Signal 2020; 13:eaba4200. [PMID: 32291314 PMCID: PMC7315801 DOI: 10.1126/scisignal.aba4200] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Breast and mammary epithelial cells experience different local environments during tissue development and tumorigenesis. Microenvironmental heterogeneity gives rise to distinct cell regulatory states whose identity and importance are just beginning to be appreciated. Cellular states diversify when clonal three-dimensional (3D) spheroids are cultured in basement membrane, and one such state is associated with stress tolerance and poor response to anticancer therapeutics. Here, we found that this state was jointly coordinated by the NRF2 and p53 pathways, which were costabilized by spontaneous oxidative stress within 3D cultures. Inhibition of NRF2 or p53 individually disrupted some of the transcripts defining the regulatory state but did not yield a notable phenotype in nontransformed breast epithelial cells. In contrast, combined perturbation prevented 3D growth in an oxidative stress-dependent manner. By integrating systems models of NRF2 and p53 signaling in a single oxidative stress network, we recapitulated these observations and made predictions about oxidative stress profiles during 3D growth. NRF2 and p53 signaling were similarly coordinated in normal breast epithelial tissue and hormone-negative ductal carcinoma in situ lesions but were uncoupled in triple-negative breast cancer (TNBC), a subtype in which p53 is usually mutated. Using the integrated model, we correlated the extent of this uncoupling in TNBC cell lines with the importance of NRF2 in the 3D growth of these cell lines and their predicted handling of oxidative stress. Our results point to an oxidative stress tolerance network that is important for single cells during glandular development and the early stages of breast cancer.
Collapse
Affiliation(s)
- Elizabeth J Pereira
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Joseph S Burns
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Christina Y Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Taylor Marohl
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Delia Calderon
- Biology and Chemistry Programs, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kristen A Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Chun-Chao Wang
- Institute of Molecular Medicine and Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
40
|
Rosenbluth JM, Schackmann RCJ, Gray GK, Selfors LM, Li CMC, Boedicker M, Kuiken HJ, Richardson A, Brock J, Garber J, Dillon D, Sachs N, Clevers H, Brugge JS. Organoid cultures from normal and cancer-prone human breast tissues preserve complex epithelial lineages. Nat Commun 2020; 11:1711. [PMID: 32249764 PMCID: PMC7136203 DOI: 10.1038/s41467-020-15548-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Recently, organoid technology has been used to generate a large repository of breast cancer organoids. Here we present an extensive evaluation of the ability of organoid culture technology to preserve complex stem/progenitor and differentiated cell types via long-term propagation of normal human mammary tissues. Basal/stem and luminal progenitor cells can differentiate in culture to generate mature basal and luminal cell types, including ER+ cells that have been challenging to maintain in culture. Cells associated with increased cancer risk can also be propagated. Single-cell analyses of matched organoid cultures and native tissues by mass cytometry for 38 markers provide a higher resolution representation of the multiple mammary epithelial cell types in the organoids, and demonstrate that protein expression patterns of the tissue of origin can be preserved in culture. These studies indicate that organoid cultures provide a valuable platform for studies of mammary differentiation, transformation, and breast cancer risk. Organoid technology has enabled the generation of several breast cancer organoids. Here, the authors combine propagation of normal human mammary tissues with mass cytometry to evaluate the ability of organoid culture technologies to preserve stem cells and differentiated cell types.
Collapse
Affiliation(s)
- Jennifer M Rosenbluth
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Ron C J Schackmann
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Carman Man-Chung Li
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Mackenzie Boedicker
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Hendrik J Kuiken
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Andrea Richardson
- Department of Pathology, Brigham & Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Jane Brock
- Department of Pathology, Brigham & Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Judy Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02115, USA
| | - Deborah Dillon
- Department of Pathology, Brigham & Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Norman Sachs
- Hubrecht Institute, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Testa U, Castelli G, Pelosi E. Breast Cancer: A Molecularly Heterogenous Disease Needing Subtype-Specific Treatments. Med Sci (Basel) 2020; 8:E18. [PMID: 32210163 PMCID: PMC7151639 DOI: 10.3390/medsci8010018] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/23/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly occurring cancer in women. There were over two-million new cases in world in 2018. It is the second leading cause of death from cancer in western countries. At the molecular level, breast cancer is a heterogeneous disease, which is characterized by high genomic instability evidenced by somatic gene mutations, copy number alterations, and chromosome structural rearrangements. The genomic instability is caused by defects in DNA damage repair, transcription, DNA replication, telomere maintenance and mitotic chromosome segregation. According to molecular features, breast cancers are subdivided in subtypes, according to activation of hormone receptors (estrogen receptor and progesterone receptor), of human epidermal growth factors receptor 2 (HER2), and or BRCA mutations. In-depth analyses of the molecular features of primary and metastatic breast cancer have shown the great heterogeneity of genetic alterations and their clonal evolution during disease development. These studies have contributed to identify a repertoire of numerous disease-causing genes that are altered through different mutational processes. While early-stage breast cancer is a curable disease in about 70% of patients, advanced breast cancer is largely incurable. However, molecular studies have contributed to develop new therapeutic approaches targeting HER2, CDK4/6, PI3K, or involving poly(ADP-ribose) polymerase inhibitors for BRCA mutation carriers and immunotherapy.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
42
|
Sumbal J, Chiche A, Charifou E, Koledova Z, Li H. Primary Mammary Organoid Model of Lactation and Involution. Front Cell Dev Biol 2020; 8:68. [PMID: 32266252 PMCID: PMC7098375 DOI: 10.3389/fcell.2020.00068] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
Mammary gland development occurs mainly after birth and is composed of three successive stages: puberty, pregnancy and lactation, and involution. These developmental stages are associated with major tissue remodeling, including extensive changes in mammary epithelium, as well as surrounding stroma. Three-dimensional (3D) mammary organoid culture has become an important tool in mammary gland biology and enabled invaluable discoveries on pubertal mammary branching morphogenesis and breast cancer. However, a suitable 3D organoid model recapitulating key aspects of lactation and involution has been missing. Here, we describe a robust and straightforward mouse mammary organoid system modeling lactation and involution-like process, which can be applied to study mechanisms of physiological mammary gland lactation and involution as well as pregnancy-associated breast cancer.
Collapse
Affiliation(s)
- Jakub Sumbal
- Department of Developmental and Stem Cell Biology, Cellular Plasticity and Disease Modelling, CNRS UMR 3738, Institut Pasteur, Paris, France
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Aurelie Chiche
- Department of Developmental and Stem Cell Biology, Cellular Plasticity and Disease Modelling, CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Elsa Charifou
- Department of Developmental and Stem Cell Biology, Cellular Plasticity and Disease Modelling, CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Han Li
- Department of Developmental and Stem Cell Biology, Cellular Plasticity and Disease Modelling, CNRS UMR 3738, Institut Pasteur, Paris, France
| |
Collapse
|
43
|
Engineering human organoid development ex vivo—challenges and opportunities. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020. [DOI: 10.1016/j.cobme.2020.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Stewart TA, Davis FM. A Primary Cell and Organoid Platform for Evaluating Pharmacological Responses in Mammary Epithelial Cells. ACS Pharmacol Transl Sci 2020; 3:63-75. [PMID: 32259089 PMCID: PMC7088941 DOI: 10.1021/acsptsci.9b00090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 12/31/2022]
Abstract
An essential process in predicting the in vivo pharmacological activity of a candidate molecule involves the evaluation of target responses using established model systems. While these models largely comprise immortalized cells, which are often serially passaged as monolayers on uniformly stiff substrates and are modified to overexpress one or more components of the pathway-of-interest, the importance of cell identity, heterogeneity, and three-dimensional (3D) context to target response is gaining increasing attention. Here, we assess intracellular calcium responses in mouse mammary epithelial cells in three distinct model systems: 3D primary organoids, 2D primary epithelial cells, and 2D immortalized cells. Specifically, we assess intracellular calcium responses to a number of extracellular signals implicated in the regulation of basal (or myoepithelial) cell function. These findings provide further insights into cell type and context-specific pharmacological responses in mammary epithelial cells and highlight the opportunities and challenges in the adoption of architecturally complex and heterogeneous in vitro assays in pharmacological research.
Collapse
Affiliation(s)
- Teneale A. Stewart
- Mater
Research-The University of Queensland, Faculty of Medicine, Brisbane, Queensland 4102, Australia
- Translational
Research Institute, Brisbane, Queensland 4102, Australia
| | - Felicity M. Davis
- Mater
Research-The University of Queensland, Faculty of Medicine, Brisbane, Queensland 4102, Australia
- Translational
Research Institute, Brisbane, Queensland 4102, Australia
| |
Collapse
|
45
|
Tang C, van den Bijgaart RJ, Looman MW, Tel-Karthaus N, de Graaf AM, Gilfillan S, Colonna M, Ansems M, Adema GJ. DC-SCRIPT deficiency delays mouse mammary gland development and branching morphogenesis. Dev Biol 2019; 455:42-50. [DOI: 10.1016/j.ydbio.2019.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 01/09/2023]
|
46
|
Fu NY, Nolan E, Lindeman GJ, Visvader JE. Stem Cells and the Differentiation Hierarchy in Mammary Gland Development. Physiol Rev 2019; 100:489-523. [PMID: 31539305 DOI: 10.1152/physrev.00040.2018] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.
Collapse
Affiliation(s)
- Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Nolan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
47
|
Slepicka PF, Cyrill SL, Dos Santos CO. Pregnancy and Breast Cancer: Pathways to Understand Risk and Prevention. Trends Mol Med 2019; 25:866-881. [PMID: 31383623 DOI: 10.1016/j.molmed.2019.06.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.
Collapse
Affiliation(s)
- Priscila F Slepicka
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
48
|
Chatterjee S, Bhat V, Berdnikov A, Liu J, Zhang G, Buchel E, Safneck J, Marshall AJ, Murphy LC, Postovit LM, Raouf A. Paracrine Crosstalk between Fibroblasts and ER + Breast Cancer Cells Creates an IL1β-Enriched Niche that Promotes Tumor Growth. iScience 2019; 19:388-401. [PMID: 31419632 PMCID: PMC6706609 DOI: 10.1016/j.isci.2019.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/16/2019] [Accepted: 07/18/2019] [Indexed: 12/30/2022] Open
Abstract
Breast cancer-induced activated fibroblasts support tumor progression. However, the role of normal fibroblasts in tumor progression remains controversial. In this study, we used modified patient-derived organoid cultures and demonstrate that constitutively secreted cytokines from normal breast fibroblasts initiate a paracrine signaling mechanism with estrogen receptor-positive (ER+) breast cancer cells, which results in the creation of an interleukin (IL)-1β-enriched microenvironment. We found that this paracrine signaling mechanism is shared between normal and activated fibroblasts. Interestingly, we observed that in reconstructed tumor microenvironment containing autologous ER+ breast cancer cells, activated fibroblasts, and immune cells, tamoxifen is more effective in reducing tumor cell proliferation when this paracrine signaling is blocked. Our findings then suggest that ER+ tumor cells could create a growth-promoting environment without activating stromal fibroblasts and that in breast-conserving surgeries, normal fibroblasts could be a significant modulator of tumor recurrence by enhancing the proliferation of residual breast cancer cells in the tumor-adjacent breast tissue. Normal fibroblast-cancer cell interaction promotes tumor progression Paracrine signaling common to normal and activated fibroblasts promotes drug resistance Fibroblast-secreted factors create an IL1β-enriched niche for ER+ breast cancer cell growth
Collapse
Affiliation(s)
- Sumanta Chatterjee
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada
| | - Vasudeva Bhat
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada
| | - Alexei Berdnikov
- Department of Surgery, Section of Plastic Surgery, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1M5, Canada
| | - Jiahui Liu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Guihua Zhang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edward Buchel
- Department of Surgery, Section of Plastic Surgery, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1M5, Canada
| | - Janice Safneck
- Department of Pathology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| | - Aaron J Marshall
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Leigh C Murphy
- Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada; Department of Biochemistry and Medical Genetics, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Obstetrics and Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Afshin Raouf
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada.
| |
Collapse
|
49
|
Dekkers JF, Alieva M, Wellens LM, Ariese HCR, Jamieson PR, Vonk AM, Amatngalim GD, Hu H, Oost KC, Snippert HJG, Beekman JM, Wehrens EJ, Visvader JE, Clevers H, Rios AC. High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc 2019; 14:1756-1771. [PMID: 31053799 DOI: 10.1038/s41596-019-0160-8] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
In vitro 3D organoid systems have revolutionized the modeling of organ development and diseases in a dish. Fluorescence microscopy has contributed to the characterization of the cellular composition of organoids and demonstrated organoids' phenotypic resemblance to their original tissues. Here, we provide a detailed protocol for performing high-resolution 3D imaging of entire organoids harboring fluorescence reporters and upon immunolabeling. This method is applicable to a wide range of organoids of differing origins and of various sizes and shapes. We have successfully used it on human airway, colon, kidney, liver and breast tumor organoids, as well as on mouse mammary gland organoids. It includes a simple clearing method utilizing a homemade fructose-glycerol clearing agent that captures 3D organoids in full and enables marker quantification on a cell-by-cell basis. Sample preparation has been optimized for 3D imaging by confocal, super-resolution confocal, multiphoton and light-sheet microscopy. From organoid harvest to image analysis, the protocol takes 3 d.
Collapse
Affiliation(s)
- Johanna F Dekkers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Maria Alieva
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Lianne M Wellens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Hendrikus C R Ariese
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Paul R Jamieson
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Annelotte M Vonk
- Regenerative Medicine Center Utrecht, University Medical Center, Utrecht University, Utrecht, the Netherlands.,Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, the Netherlands
| | - Gimano D Amatngalim
- Regenerative Medicine Center Utrecht, University Medical Center, Utrecht University, Utrecht, the Netherlands.,Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, the Netherlands
| | - Huili Hu
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Koen C Oost
- Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hugo J G Snippert
- Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeffrey M Beekman
- Regenerative Medicine Center Utrecht, University Medical Center, Utrecht University, Utrecht, the Netherlands.,Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, the Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Jane E Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands.,Cancer Genomics Center (CGC), Utrecht, the Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands. .,Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht, the Netherlands. .,Cancer Genomics Center (CGC), Utrecht, the Netherlands.
| |
Collapse
|
50
|
Yoo KH, Hennighausen L, Shin HY. Dissecting Tissue-Specific Super-Enhancers by Integrating Genome-Wide Analyses and CRISPR/Cas9 Genome Editing. J Mammary Gland Biol Neoplasia 2019; 24:47-59. [PMID: 30291498 DOI: 10.1007/s10911-018-9417-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022] Open
Abstract
Recent advances in genome-wide sequencing technologies have provided researchers with unprecedented opportunities to discover the genomic structures of gene regulatory units in living organisms. In particular, the integration of ChIP-seq, RNA-seq, and DNase-seq techniques has facilitated the mapping of a new class of regulatory elements. These elements, called super-enhancers, can regulate cell-type-specific gene sets and even fine-tune gene expression regulation in response to external stimuli, and have become a hot topic in genome biology. However, there is scant genetic evidence demonstrating their unique biological relevance and the mechanisms underlying these biological functions. In this review, we describe a robust genome-wide strategy for mapping cell-type-specific enhancers or super-enhancers in the mammary genome. In this strategy, genome-wide screening of active enhancer clusters that are co-occupied by mammary-enriched transcription factors, co-factors, and active enhancer marks is used to identify bona fide mammary tissue-specific super-enhancers. The in vivo function of these super-enhancers and their associated regulatory elements may then be investigated in various ways using the advanced CRISPR/Cas9 genome-editing technology. Based on our experience targeting various mammary genomic sites using CRISPR/Cas9 in mice, we comprehensively discuss the molecular consequences of the different targeting methods, such as the number of gRNAs and the dependence on their simultaneous or sequential injections. We also mention the considerations that are essential for obtaining accurate results and shed light on recent progress that has been made in developing modified CRISPR/Cas9 genome-editing techniques. In the future, the coupling of advanced genome-wide sequencing and genome-editing technologies could provide new insights into the complex genetic regulatory networks involved in mammary-gland development.
Collapse
Affiliation(s)
- Kyung Hyun Yoo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institutes of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- BK21 Biological Science Visiting Professor, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ha Youn Shin
- Department of Biomedical Science and Engineering, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|