1
|
Johnstone M, Leck A, Lange T, Wilcher K, Shephard MS, Paranjpe A, Schutte S, Wells S, Kappes F, Salomonis N, Privette Vinnedge LM. The chromatin remodeler DEK promotes proliferation of mammary epithelium and is associated with H3K27me3 epigenetic modifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612116. [PMID: 39314335 PMCID: PMC11419013 DOI: 10.1101/2024.09.09.612116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The DEK chromatin remodeling protein was previously shown to confer oncogenic phenotypes to human and mouse mammary epithelial cells using in vitro and knockout mouse models. However, its functional role in normal mammary gland epithelium remained unexplored. We developed two novel mouse models to study the role of Dek in normal mammary gland biology in vivo . Mammary gland-specific Dek over-expression in mice resulted in hyperproliferation of cells that visually resembled alveolar cells, and a transcriptional profile that indicated increased expression of cell cycle, mammary stem/progenitor, and lactation-associated genes. Conversely, Dek knockout mice exhibited an alveologenesis or lactation defect, resulting in dramatically reduced pup survival. Analysis of previously published single-cell RNA-sequencing of mouse mammary glands revealed that Dek is most highly expressed in mammary stem cells and alveolar progenitor cells, and to a lesser extent in basal epithelial cells, supporting the observed phenotypes. Mechanistically, we discovered that Dek is a modifier of Ezh2 methyltransferase activity, upregulating the levels of histone H3 trimethylation on lysine 27 (H3K27me3) to control gene transcription. Combined, this work indicates that Dek promotes proliferation of mammary epithelial cells via cell cycle deregulation. Furthermore, we report a novel function for Dek in alveologenesis and histone H3 K27 trimethylation.
Collapse
|
2
|
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
|
3
|
Khan IR, Sadida HQ, Hashem S, Singh M, Macha MA, Al-Shabeeb Akil AS, Khurshid I, Bhat AA. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed Pharmacother 2024; 176:116873. [PMID: 38843587 DOI: 10.1016/j.biopha.2024.116873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Esophageal cancer (EC) is significantly influenced by the tumor microenvironment (TME) and altered signaling pathways. Downregulating these pathways in EC is essential for suppressing tumor development, preventing metastasis, and enhancing therapeutic outcomes. This approach can increase tumor sensitivity to treatments, enhance patient outcomes, and inhibit cancer cell proliferation and spread. The TME, comprising cellular and non-cellular elements surrounding the tumor, significantly influences EC's development, course, and treatment responsiveness. Understanding the complex relationships within the TME is crucial for developing successful EC treatments. Immunotherapy is a vital TME treatment for EC. However, the heterogeneity within the TME limits the application of anticancer drugs outside clinical settings. Therefore, identifying reliable microenvironmental biomarkers that can detect therapeutic responses before initiating therapy is crucial. Combining approaches focusing on EC signaling pathways with TME can enhance treatment outcomes. This integrated strategy aims to interfere with essential signaling pathways promoting cancer spread while disrupting factors encouraging tumor development. Unraveling aberrant signaling pathways and TME components can lead to more focused and efficient treatment approaches, identifying specific cellular targets for treatments. Targeting the TME and signaling pathways may reduce metastasis risk by interfering with mechanisms facilitating cancer cell invasion and dissemination. In conclusion, this integrative strategy has significant potential for improving patient outcomes and advancing EC research and therapy. This review discusses the altered signaling pathways and TME in EC, focusing on potential future therapeutics.
Collapse
Affiliation(s)
- Inamu Rashid Khan
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine Doha 26999, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir 192122, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Ibraq Khurshid
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar.
| |
Collapse
|
4
|
Koskinen LM, Nieminen L, Arjonen A, Guzmán C, Peurla M, Peuhu E. Spatial Engineering of Mammary Epithelial Cell Cultures with 3D Bioprinting Reveals Growth Control by Branch Point Proximity. J Mammary Gland Biol Neoplasia 2024; 29:5. [PMID: 38416267 PMCID: PMC10902034 DOI: 10.1007/s10911-024-09557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
The three-dimensional (3D) structure of the ductal epithelium and the surrounding extracellular matrix (ECM) are integral aspects of the breast tissue, and they have important roles during mammary gland development, function and malignancy. However, the architecture of the branched mammary epithelial network is poorly recapitulated in the current in vitro models. 3D bioprinting is an emerging approach to improve tissue-mimicry in cell culture. Here, we developed and optimized a protocol for 3D bioprinting of normal and cancerous mammary epithelial cells into a branched Y-shape to study the role of cell positioning in the regulation of cell proliferation and invasion. Non-cancerous cells formed continuous 3D cell networks with several organotypic features, whereas the ductal carcinoma in situ (DCIS) -like cancer cells exhibited aberrant basal polarization and defective formation of the basement membrane (BM). Quantitative analysis over time demonstrated that both normal and cancerous cells proliferate more at the branch tips compared to the trunk region of the 3D-bioprinted cultures, and particularly at the tip further away from the branch point. The location-specific rate of proliferation was independent of TGFβ signaling but invasion of the DCIS-like breast cancer cells was reduced upon the inhibition of TGFβ. Thus, our data demonstrate that the 3D-bioprinted cells can sense their position in the branched network of cells and proliferate at the tips, thus recapitulating this feature of mammary epithelial branching morphogenesis. In all, our results demonstrate the capacity of the developed 3D bioprinting method for quantitative analysis of the relationships between tissue structure and cell behavior in breast morphogenesis and cancer.
Collapse
Affiliation(s)
- Leena M Koskinen
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | | | | - Markus Peurla
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
5
|
Tomasin R, Rodrigues AM, Manucci AC, Bruni-Cardoso A. A molecular landscape of quiescence and proliferation highlights the role of Pten in mammary gland acinogenesis. J Cell Sci 2023; 136:jcs261178. [PMID: 37712332 DOI: 10.1242/jcs.261178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023] Open
Abstract
Cell context is key for cell state. Using physiologically relevant models of laminin-rich extracellular matrix (lrECM) induction of mammary epithelial cell quiescence and differentiation, we provide a landscape of the key molecules for the proliferation-quiescence decision, identifying multiple layers of regulation at the mRNA and protein levels. Quiescence occurred despite activity of Fak (also known as PTK2), Src and phosphoinositide 3-kinases (PI3Ks), suggesting the existence of a disconnecting node between upstream and downstream proliferative signalling. Pten, a lipid and protein phosphatase, fulfils this role, because its inhibition increased proliferation and restored signalling via the Akt, mTORC1, mTORC2 and mitogen-activated protein kinase (MAPK) pathways. Pten and laminin levels were positively correlated in developing murine mammary epithelia, and Pten localized apicolaterally in luminal cells in ducts and near the nascent lumen in terminal end buds. Consistently, in three-dimensional acinogenesis models, Pten was required for triggering and sustaining quiescence, polarity and architecture. The multilayered regulatory circuitry that we uncovered provides an explanation for the robustness of quiescence within a growth-suppressive microenvironment, which could nonetheless be disrupted by perturbations in master regulators such as Pten.
Collapse
Affiliation(s)
- Rebeka Tomasin
- E-signal lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Ana Maria Rodrigues
- E-signal lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Antonio Carlos Manucci
- E-signal lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Alexandre Bruni-Cardoso
- E-signal lab, Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
6
|
Daneshdoust D, Luo M, Li Z, Mo X, Alothman S, Kallakury B, Schlegel R, Zhang J, Guo D, Furth PA, Liu X, Li J. Unlocking Translational Potential: Conditionally Reprogrammed Cells in Advancing Breast Cancer Research. Cells 2023; 12:2388. [PMID: 37830602 PMCID: PMC10572051 DOI: 10.3390/cells12192388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Preclinical in vitro models play an important role in studying cancer cell biology and facilitating translational research, especially in the identification of drug targets and drug discovery studies. This is particularly relevant in breast cancer, where the global burden of disease is quite high based on prevalence and a relatively high rate of lethality. Predictive tools to select patients who will be responsive to invasive or morbid therapies (radiotherapy, chemotherapy, immunotherapy, and/or surgery) are relatively lacking. To be clinically relevant, a model must accurately replicate the biology and cellular heterogeneity of the primary tumor. Addressing these requirements and overcoming the limitations of most existing cancer cell lines, which are typically derived from a single clone, we have recently developed conditional reprogramming (CR) technology. The CR technology refers to a co-culture system of primary human normal or tumor cells with irradiated murine fibroblasts in the presence of a Rho-associated kinase inhibitor to allow the primary cells to acquire stem cell properties and the ability to proliferate indefinitely in vitro without any exogenous gene or viral transfection. This innovative approach fulfills many of these needs and offers an alternative that surpasses the deficiencies associated with traditional cancer cell lines. These CR cells (CRCs) can be reprogrammed to maintain a highly proliferative state and reproduce the genomic and histological characteristics of the parental tissue. Therefore, CR technology may be a clinically relevant model to test and predict drug sensitivity, conduct gene profile analysis and xenograft research, and undertake personalized medicine. This review discusses studies that have applied CR technology to conduct breast cancer research.
Collapse
Affiliation(s)
- Danyal Daneshdoust
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Mingjue Luo
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Zaibo Li
- Departments of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Department of Biostatics and Bioinformatics, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Sahar Alothman
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Bhaskar Kallakury
- Departments of Pathology, Lombardi Comprehensive Cancer Center, Center for Cell Reprogramming, Georgetown University, Washington, DC 20057, USA
| | - Richard Schlegel
- Departments of Pathology, Lombardi Comprehensive Cancer Center, Center for Cell Reprogramming, Georgetown University, Washington, DC 20057, USA
| | - Junran Zhang
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Priscilla A. Furth
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Departments of Pathology, Urology, and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Jenny Li
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Moody J, Mears E, Trevarton AJ, Broadhurst M, Molenaar A, Chometon T, Lopdell T, Littlejohn M, Snell R. Successful editing and maintenance of lactogenic gene expression in primary bovine mammary epithelial cells. In Vitro Cell Dev Biol Anim 2023:10.1007/s11626-023-00762-6. [PMID: 37278965 DOI: 10.1007/s11626-023-00762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/23/2023] [Indexed: 06/07/2023]
Abstract
In vitro investigation of bovine lactation processes is limited by a lack of physiologically representative cell models. This deficiency is most evident through the minimal or absent expression of lactation-specific genes in cultured bovine mammary tissues. Primary bovine mammary epithelial cells (pbMECs) extracted from lactating mammary tissue and grown in culture initially express milk protein transcripts at relatively representative levels. However, expression drops dramatically after only three or four passages, which greatly reduces the utility of primary cells to model and further examine lactogenesis. To investigate the effects of alternate alleles in pbMECs including effects on transcription, we have developed methods to deliver CRISPR-Cas9 gene editing reagents to primary mammary cells, resulting in very high editing efficiencies. We have also found that culturing the cells on an imitation basement membrane composed of Matrigel, results in the restoration of a more representative lactogenic gene expression profile and the cells forming three-dimensional structures in vitro. Here, we present data from four pbMEC lines recovered from pregnant cows and detail the expression profile of five key milk synthesis genes in these MECs grown on Matrigel. Additionally, we describe an optimised method for preferentially selecting CRISPR-Cas9-edited cells conferring a knock-out of DGAT1, using fluorescence-activated cell sorting (FACS). The combination of these techniques facilitates the use of pbMECs as a model to investigate the effects of gene introgressions and genetic variation in lactating mammary tissue.
Collapse
Affiliation(s)
- Janelle Moody
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand.
| | - Emily Mears
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Alexander J Trevarton
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | | - Thaize Chometon
- Faculty of Sciences, Auckland Cytometry, The University of Auckland, Auckland, New Zealand
| | - Thomas Lopdell
- Livestock Improvement Corporation, Hamilton, New Zealand
| | | | - Russell Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
Feitor JF, Brazaca LC, Lima AM, Ferreira VG, Kassab G, Bagnato VS, Carrilho E, Cardoso DR. Organ-on-a-Chip for Drug Screening: A Bright Future for Sustainability? A Critical Review. ACS Biomater Sci Eng 2023; 9:2220-2234. [PMID: 37014814 DOI: 10.1021/acsbiomaterials.2c01454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Globalization has raised concerns about spreading diseases and emphasized the need for quick and efficient methods for drug screening. Established drug efficacy and toxicity approaches have proven obsolete, with a high failure rate in clinical trials. Organ-on-a-chip has emerged as an essential alternative to outdated techniques, precisely simulating important characteristics of organs and predicting drug pharmacokinetics more ethically and efficiently. Although promising, most organ-on-a-chip devices are still manufactured using principles and materials from the micromachining industry. The abusive use of plastic for traditional drug screening methods and device production should be considered when substituting technologies so that the compensation for the generation of plastic waste can be projected. This critical review outlines recent advances for organ-on-a-chip in the industry and estimates the possibility of scaling up its production. Moreover, it analyzes trends in organ-on-a-chip publications and provides suggestions for a more sustainable future for organ-on-a-chip research and production.
Collapse
Affiliation(s)
- Jéssica F Feitor
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Laís C Brazaca
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, 02138 Massachusetts, United States
| | - Amanda M Lima
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Vinícius G Ferreira
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Giulia Kassab
- Instituto de Física de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Vanderlei S Bagnato
- Instituto de Física de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica-INCTBio, 13083-970 Campinas, SP, Brazil
| | - Daniel R Cardoso
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| |
Collapse
|
9
|
Zhou Y, Ye Z, Wei W, Zhang M, Huang F, Li J, Cai C. Macrophages maintain mammary stem cell activity and mammary homeostasis via TNF-α-PI3K-Cdk1/Cyclin B1 axis. NPJ Regen Med 2023; 8:23. [PMID: 37130846 PMCID: PMC10154328 DOI: 10.1038/s41536-023-00296-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/20/2023] [Indexed: 05/04/2023] Open
Abstract
Adult stem cell niche is a special environment composed of a variety stromal cells and signals, which cooperatively regulate tissue development and homeostasis. It is of great interest to study the role of immune cells in niche. Here, we show that mammary resident macrophages regulate mammary epithelium cell division and mammary development through TNF-α-Cdk1/Cyclin B1 axis. In vivo, depletion of macrophages reduces the number of mammary basal cells and mammary stem cells (MaSCs), while increases mammary luminal cells. In vitro, we establish a three-dimensional culture system in which mammary basal cells are co-cultured with macrophages, and interestingly, macrophage co-culture promotes the formation of branched functional mammary organoids. Moreover, TNF-α produced by macrophages activates the intracellular PI3K/Cdk1/Cyclin B1 signaling in mammary cells, thereby maintaining the activity of MaSCs and the formation of mammary organoids. Together, these findings reveal the functional significance of macrophageal niche and intracellular PI3K/Cdk1/Cyclin B1 axis for maintaining MaSC activity and mammary homeostasis.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zi Ye
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengna Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fujing Huang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
10
|
Alshehri S, Pavlovič T, Farsinejad S, Behboodi P, Quan L, Centeno D, Kung D, Rezler M, Lee W, Jasiński P, Dziabaszewska E, Nowak-Markwitz E, Kalyon D, Zaborowski MP, Iwanicki M. Extracellular Matrix Modulates Outgrowth Dynamics in Ovarian Cancer. Adv Biol (Weinh) 2022; 6:e2200197. [PMID: 36084257 PMCID: PMC9772079 DOI: 10.1002/adbi.202200197] [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: 07/15/2022] [Revised: 08/18/2022] [Indexed: 01/28/2023]
Abstract
Ovarian carcinoma (OC) forms outgrowths that extend from the outer surface of an afflicted organ into the peritoneum. OC outgrowth formation is poorly understood due to the limited availability of cell culture models examining the behavior of cells that form outgrowths. Prompted by immunochemical evaluation of extracellular matrix (ECM) components in human tissues, laminin and collagen-rich ECM-reconstituted cell culture models amenable to studies of cell clusters that can form outgrowths are developed. It is demonstrated that ECM promotes outgrowth formation in fallopian tube non-ciliated epithelial cells (FNE) expressing mutant p53 and various OC cell lines. Outgrowths are initiated by cells that underwent outward translocation and retained the ability to intercalate into mesothelial cell monolayers. Electron microscopy, optical coherence tomography, and small amplitude oscillatory shear experiments reveal that increased ECM levels led to increased fibrous network thickness and high shear elasticity of the microenvironment. These physical characteristics are associated with outgrowth suppression. The low ECM microenvironment mimicks the viscoelasticity of malignant peritoneal fluid (ascites) and supports cell proliferation, cell translocation, and outgrowth formation. These results highlight the importance of the ECM microenvironment in modulating OC growth and can provide additional insights into the mode of dissemination of primary and recurrent ovarian tumors.
Collapse
Affiliation(s)
- Sarah Alshehri
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Tonja Pavlovič
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Sadaf Farsinejad
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Panteha Behboodi
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Li Quan
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Daniel Centeno
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Douglas Kung
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Marta Rezler
- Poznań University of Medical Sciences, Collegium Maius, Fredry 10, Poznań, 61-701, Poland
| | - Woo Lee
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Piotr Jasiński
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Elżbieta Dziabaszewska
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Dilhan Kalyon
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Mikołaj P Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Marcin Iwanicki
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| |
Collapse
|
11
|
Vigili de Kreutzenberg S, Giannella A, Ceolotto G, Faggin E, Cappellari R, Mazzucato M, Fraccaro C, Tarantini G, Avogaro A, Fadini GP. A miR-125/Sirtuin-7 pathway drives the pro-calcific potential of myeloid cells in diabetic vascular disease. Diabetologia 2022; 65:1555-1568. [PMID: 35708762 PMCID: PMC9345831 DOI: 10.1007/s00125-022-05733-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/22/2022] [Indexed: 11/03/2022]
Abstract
AIMS/HYPOTHESIS Ectopic calcification is a typical feature of diabetic vascular disease and resembles an accelerated ageing phenotype. We previously found an excess of myeloid calcifying cells in diabetic individuals. We herein examined molecular and cellular pathways linking atherosclerotic calcification with calcification by myeloid cells in the diabetic milieu. METHODS We first examined the associations among coronary calcification, myeloid calcifying cell levels and mononuclear cell gene expression in a cross-sectional study of 87 participants with type 2 diabetes undergoing elective coronary angiography. Then, we undertook in vitro studies on mesenchymal stem cells and the THP-1 myeloid cell line to verify the causal relationships of the observed associations. RESULTS Coronary calcification was associated with 2.8-times-higher myeloid calcifying cell levels (p=0.037) and 50% elevated expression of the osteogenic gene RUNX2 in mononuclear cells, whereas expression of Sirtuin-7 (SIRT7) was inversely correlated with calcification. In standard differentiation assays of mesenchymal stem cells, SIRT7 knockdown activated the osteogenic program and worsened calcification, especially in the presence of high (20 mmol/l) glucose. In the myeloid cell line THP-1, SIRT7 downregulation drove a pro-calcific phenotype, whereas SIRT7 overexpression prevented high-glucose-induced calcification. Through the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, high glucose induced miR-125b-5p, which in turn targeted SIRT7 in myeloid cells and was directly associated with coronary calcification. CONCLUSIONS/INTERPRETATION We describe a new pathway elicited by high glucose through the JAK/STAT cascade, involving regulation of SIRT7 by miR-125b-5p and driving calcification by myeloid cells. This pathway is associated with coronary calcification in diabetic individuals and may be a target against diabetic vascular disease. DATA AVAILABILITY RNA sequencing data are deposited in GEO (accession number GSE193510; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE193510 ).
Collapse
Affiliation(s)
| | | | - Giulio Ceolotto
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | | | - Roberta Cappellari
- Department of Medicine - DIMED, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marta Mazzucato
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Chiara Fraccaro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Tarantini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine - DIMED, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
12
|
Selma-Royo M, Calvo-Lerma J, Bäuerl C, Esteban-Torres M, Cabrera-Rubio R, Collado MC. Human milk microbiota: what did we learn in the last 20 years? MICROBIOME RESEARCH REPORTS 2022; 1:19. [PMID: 38046359 PMCID: PMC10688795 DOI: 10.20517/mrr.2022.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/05/2023]
Abstract
Human milk (HM) is the gold standard for infant nutrition during the first months of life. Beyond its nutritional components, its complex bioactive composition includes microorganisms, their metabolites, and oligosaccharides, which also contribute to gut colonization and immune system maturation. There is growing evidence of the beneficial effects of bacteria present in HM. However, current research presents limited data on the presence and functions of other organisms. The potential biological impacts on maternal and infant health outcomes, the factors contributing to milk microbes' variations, and the potential functions in the infant's gut remain unclear. This review provides a global overview of milk microbiota, what the actual knowledge is, and what the gaps and challenges are for the next years.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia 46980, Spain
| |
Collapse
|
13
|
Seitlinger J, Nounsi A, Idoux-Gillet Y, Santos Pujol E, Lê H, Grandgirard E, Olland A, Lindner V, Zaupa C, Balloul JM, Quemeneur E, Massard G, Falcoz PE, Hua G, Benkirane-Jessel N. Vascularization of Patient-Derived Tumoroid from Non-Small-Cell Lung Cancer and Its Microenvironment. Biomedicines 2022; 10:biomedicines10051103. [PMID: 35625840 PMCID: PMC9138465 DOI: 10.3390/biomedicines10051103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Patient-derived tumoroid (PDT) has been developed and used for anti-drug screening in the last decade. As compared to other existing drug screening models, a PDT-based in vitro 3D cell culture model could preserve the histological and mutational characteristics of their corresponding tumors and mimic the tumor microenvironment. However, few studies have been carried out to improve the microvascular network connecting the PDT and its surrounding microenvironment, knowing that poor tumor-selective drug transport and delivery is one of the major reasons for both the failure of anti-cancer drug screens and resistance in clinical treatment. In this study, we formed vascularized PDTs in six days using multiple cell types which maintain the histopathological features of the original cancer tissue. Furthermore, our results demonstrated a vascular network connecting PDT and its surrounding microenvironment. This fast and promising PDT model opens new perspectives for personalized medicine: this model could easily be used to test all therapeutic treatments and could be connected with a microfluidic device for more accurate drug screening.
Collapse
Affiliation(s)
- Joseph Seitlinger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- 1 Place de l’Hôpital, University Hospital Strasbourg (HUS), 67000 Strasbourg, France
| | - Anasse Nounsi
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Eloy Santos Pujol
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Hélène Lê
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
- Transgene SA, 400 Boulevard Gonthier d’Andernach-Parc d’Innovation-CS80166, 67405 Illkirch Graffenstaden, France; (C.Z.); (J.-M.B.); (E.Q.)
| | - Erwan Grandgirard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS, UMR 7104, Inserm U 1258, 1 rue Laurent Fries, BP 10142, 67404 Illkirch Graffenstaden, France;
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- 1 Place de l’Hôpital, University Hospital Strasbourg (HUS), 67000 Strasbourg, France
| | - Véronique Lindner
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- 1 Place de l’Hôpital, University Hospital Strasbourg (HUS), 67000 Strasbourg, France
| | - Cécile Zaupa
- Transgene SA, 400 Boulevard Gonthier d’Andernach-Parc d’Innovation-CS80166, 67405 Illkirch Graffenstaden, France; (C.Z.); (J.-M.B.); (E.Q.)
| | - Jean-Marc Balloul
- Transgene SA, 400 Boulevard Gonthier d’Andernach-Parc d’Innovation-CS80166, 67405 Illkirch Graffenstaden, France; (C.Z.); (J.-M.B.); (E.Q.)
| | - Eric Quemeneur
- Transgene SA, 400 Boulevard Gonthier d’Andernach-Parc d’Innovation-CS80166, 67405 Illkirch Graffenstaden, France; (C.Z.); (J.-M.B.); (E.Q.)
| | - Gilbert Massard
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- 1 Place de l’Hôpital, University Hospital Strasbourg (HUS), 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Pierre-Emmanuel Falcoz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- 1 Place de l’Hôpital, University Hospital Strasbourg (HUS), 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Guoqiang Hua
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (J.S.); (A.N.); (Y.I.-G.); (E.S.P.); (H.L.); (A.O.); (V.L.); (G.M.); (P.-E.F.); (G.H.)
- Faculty of Dental Surgery, University of Strasbourg, 67000 Strasbourg, France
- Faculty of medicine, University of Strasbourg, 67000 Strasbourg, France
- Correspondence:
| |
Collapse
|
14
|
Cambra HM, Tallapragada NP, Mannam P, Breault DT, Klein AM. Triple-Decker Sandwich Cultures of Intestinal Organoids for Long-Term Live Imaging, Uniform Perturbation, and Statistical Sampling. Curr Protoc 2022; 2:e330. [PMID: 35030297 PMCID: PMC9006308 DOI: 10.1002/cpz1.330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Three-dimensional organoid cultures enable the study of stem cell and tissue biology ex vivo, providing improved access to cells for perturbation and live imaging. Typically, organoids are grown in hydrogel domes that are simple to prepare but that lead to non-uniform tissue growth and viability. We recently developed a simple alternative culture method to embed intestinal organoids in multilayered hydrogels, called "triple-decker sandwiches," that align organoids in a common z-plane with uniform access to medium. This culture configuration improves the growth and survival of organoids over a wide working area and facilitates long-term confocal imaging and molecular perturbation. Here, we present protocols for preparing organoids in triple-decker sandwich cultures and using them for live imaging, immunostaining, and single-cell RNA sequencing. We have tested our methods on mouse and human intestinal organoids and expect them to be useful for other highly proliferative three-dimensional cell cultures. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Pre-coating plates with PolyHEMA to prepare them for triple-decker sandwich culture Support Protocol 1: Preparing PolyHEMA solution to coat glass-bottom dishes Basic Protocol 2: Embedding intestinal organoids in triple-decker sandwiches Alternate Protocol 1: Seeding single cells or organoids at low density in triple-decker sandwiches Support Protocol 2: Embedding intestinal organoids in hydrogel domes Support Protocol 3: Production of Wnt3a-conditioned medium Support Protocol 4: Production of Rspo1-conditioned medium Basic Protocol 3: Live imaging of mouse intestinal organoids in triple-decker sandwich cultures Alternate Protocol 2: Live imaging of vital dye-treated mouse intestinal organoids in triple-decker sandwich cultures Basic Protocol 4: Immunofluorescence imaging of mouse organoids liberated from triple-decker sandwich cultures Alternate Protocol 3: Liberating and fixing mouse intestinal organoids from dome cultures Support Protocol 5: Measuring cell proliferation by EdU staining of mouse intestinal organoids Basic Protocol 5: Single-cell RNA sequencing and analysis of mouse intestinal organoids.
Collapse
Affiliation(s)
- Hailey M. Cambra
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Naren P. Tallapragada
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Prabhath Mannam
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard University, Boston, MA 02139, USA
| | - Allon M. Klein
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA,Lead contact,Correspondence:
| |
Collapse
|
15
|
Del Valle LG, Montero MG, Jechlinger M. Modification of Single Cells Within Mouse Mammary Gland Derived Acini via Viral Transduction. Methods Mol Biol 2022; 2471:185-194. [PMID: 35175597 DOI: 10.1007/978-1-0716-2193-6_9] [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
The growth of organoid cultures from primary donor tissue is able to recapitulate the original tissue morphology, heterogeneity, and characteristics. Close study of these cultures grants a deeper understanding of the chain of events occurring during disease progression and healthy tissue development. While patient derived organoids are particularly suited to assay for novel treatment options, organoids obtained from model organisms are perfectly suited to establish in-depth analysis technology, including longitudinal imaging approaches, as well as proof of principle studies that rely on a steady source of primary tissue. All these approaches profit from advancements in technology to manipulate cells within an organoid.Here we present an optimized protocol to generate, culture, and transduce 3D acini obtained from mouse primary mammary epithelial cells via viral vectors. Applying this method, a few cells within the preserved organoid can be marked, changed, and tracked within an unaltered neighboring environment of non-transduced cells to better understand processes like, for instance, tumor initiation.
Collapse
|
16
|
Rauner G, Jin DX, Miller DH, Gierahn TM, Li CM, Sokol ES, Feng YX, Mathis RA, Love JC, Gupta PB, Kuperwasser C. Breast tissue regeneration is driven by cell-matrix interactions coordinating multi-lineage stem cell differentiation through DDR1. Nat Commun 2021; 12:7116. [PMID: 34893587 PMCID: PMC8664951 DOI: 10.1038/s41467-021-27401-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/15/2021] [Indexed: 11/09/2022] Open
Abstract
Mammary morphogenesis is an orchestrated process involving differentiation, proliferation and organization of cells to form a bi-layered epithelial network of ducts and lobules embedded in stromal tissue. We have engineered a 3D biomimetic human breast that makes it possible to study how stem cell fate decisions translate to tissue-level structure and function. Using this advancement, we describe the mechanism by which breast epithelial cells build a complex three-dimensional, multi-lineage tissue by signaling through a collagen receptor. Discoidin domain receptor tyrosine kinase 1 induces stem cells to differentiate into basal cells, which in turn stimulate luminal progenitor cells via Notch signaling to differentiate and form lobules. These findings demonstrate how human breast tissue regeneration is triggered by transmission of signals from the extracellular matrix through an epithelial bilayer to coordinate structural changes that lead to formation of a complex ductal-lobular network. Mammary morphogenesis is a complex process. Here the authors describe how stem cells build a three-dimensional self-organizing multi-lineage tissue by showing that positional signals from the extracellular matrix through the collagen receptor DDR1 lead stem cells to differentiate into multi-lineage committed multi-layered progeny.
Collapse
Affiliation(s)
- Gat Rauner
- Department of Developmental, Chemical & Molecular Biology, Tufts University, Boston, MA, 02111, USA
| | - Dexter X Jin
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel H Miller
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Todd M Gierahn
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Carman M Li
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Ethan S Sokol
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yu-Xiong Feng
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Robert A Mathis
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02129, USA
| | - Piyush B Gupta
- Department of Developmental, Chemical & Molecular Biology, Tufts University, Boston, MA, 02111, USA. .,Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Laboratory for the Convergence of Biomedical, Physical, and Engineering Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
| | - Charlotte Kuperwasser
- Department of Developmental, Chemical & Molecular Biology, Tufts University, Boston, MA, 02111, USA. .,Laboratory for the Convergence of Biomedical, Physical, and Engineering Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
| |
Collapse
|
17
|
Barcellona MN, Speer JE, Jing L, Patil DS, Gupta MC, Buchowski JM, Setton LA. Bioactive in situ crosslinkable polymer-peptide hydrogel for cell delivery to the intervertebral disc in a rat model. Acta Biomater 2021; 131:117-127. [PMID: 34229105 PMCID: PMC9157564 DOI: 10.1016/j.actbio.2021.06.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022]
Abstract
Degeneration of the intervertebral disc (IVD) is associated with
significant biochemical and morphological changes that include a loss of disc
height, decreased water content and decreased cellularity. Cell delivery has
been widely explored as a strategy to supplement the nucleus pulposus (NP)
region of the degenerated IVD in both pre-clinical and clinical trials, using
progenitor or primary cell sources. We previously demonstrated an ability for a
polymer-peptide hydrogel, serving as a culture substrate, to promote adult NP
cells to undergo a shift from a degenerative fibroblast-like state to a
juvenile-like NP phenotype. In the current study, we evaluate the ability for
this peptide-functionalized hydrogel to serve as a bioactive system for cell
delivery, retention and preservation of a biosynthetic phenotype for primary IVD
cells delivered to the rat caudal disc in an anular puncture degeneration model.
Our data suggest that encapsulation of adult degenerative human NP cells in a
stiff formulation of the hydrogel functionalized with laminin-mimetic peptides
IKVAV and AG73 can promote cell viability and increased biosynthetic activity
for this population in 3D culture in vitro. Delivery of the
peptide-functionalized biomaterial with primary rat cells to the degenerated IVD
supported NP cell retention and NP-specific protein expression in
vivo, and promoted improved disc height index (DHI) values and
endplate organization compared to untreated degenerated controls. The results of
this study suggest the physical cues of this peptide-functionalized hydrogel can
serve as a supportive carrier for cell delivery to the IVD.
Collapse
Affiliation(s)
- Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, United States
| | - Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, United States
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, United States
| | - Deepanjali S Patil
- Department of Biomedical Engineering, Washington University in St. Louis, United States
| | - Munish C Gupta
- Department of Orthopedic Surgery, Washington University School of Medicine, United States
| | - Jacob M Buchowski
- Department of Orthopedic Surgery, Washington University School of Medicine, United States
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, United States; Department of Orthopedic Surgery, Washington University School of Medicine, United States.
| |
Collapse
|
18
|
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: 50] [Impact Index Per Article: 12.5] [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
|
19
|
Tallapragada NP, Cambra HM, Wald T, Keough Jalbert S, Abraham DM, Klein OD, Klein AM. Inflation-collapse dynamics drive patterning and morphogenesis in intestinal organoids. Cell Stem Cell 2021; 28:1516-1532.e14. [PMID: 33915079 DOI: 10.1016/j.stem.2021.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 12/29/2020] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
How stem cells self-organize to form structured tissues is an unsolved problem. Intestinal organoids offer a model of self-organization as they generate stem cell zones (SCZs) of typical size even without a spatially structured environment. Here we examine processes governing the size of SCZs. We improve the viability and homogeneity of intestinal organoid cultures to enable long-term time-lapse imaging of multiple organoids in parallel. We find that SCZs are shaped by fission events under strong control of ion channel-mediated inflation and mechanosensitive Piezo-family channels. Fission occurs through stereotyped modes of dynamic behavior that differ in their coordination of budding and differentiation. Imaging and single-cell transcriptomics show that inflation drives acute stem cell differentiation and induces a stretch-responsive cell state characterized by large transcriptional changes, including upregulation of Piezo1. Our results reveal an intrinsic capacity of the intestinal epithelium to self-organize by modulating and then responding to its mechanical state.
Collapse
Affiliation(s)
- Naren P Tallapragada
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hailey M Cambra
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Tomas Wald
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Samantha Keough Jalbert
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Diana M Abraham
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Allon M Klein
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Zhao X, Cong S, Guo Q, Cheng Y, Liang T, Wang J, Zhang G. Combination of Immune-Related Genomic Alterations Reveals Immune Characterization and Prediction of Different Prognostic Risks in Ovarian Cancer. Front Cell Dev Biol 2021; 9:653357. [PMID: 33968933 PMCID: PMC8102990 DOI: 10.3389/fcell.2021.653357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/17/2021] [Indexed: 11/13/2022] Open
Abstract
With the highest case-fatality rate among women, the molecular pathological alterations of ovarian cancer (OV) are complex, depending on the diversity of genomic alterations. Increasing evidence supports that immune infiltration in tumors is associated with prognosis. Therefore, we aim to assess infiltration in OV using multiple methods to capture genomic signatures regulating immune events to identify reliable predictions of different outcomes. A dataset of 309 ovarian serous cystadenocarcinoma patients with overall survival >90 days from The Cancer Genome Atlas (TCGA) was analyzed. Multiple estimations and clustering methods identified and verified two immune clusters with component differences. Functional analyses pointed out immune-related alterations underlying internal genomic variables potentially. After extracting immune genes from a public database, the LASSO Cox regression model with 10-fold cross-validation was used for selecting genes associated with overall survival rate significantly, and a risk score model was then constructed. Kaplan-Meier survival and Cox regression analyses among cohorts were performed systematically to evaluate prognostic efficiency among the risk score model and other clinical pathological parameters, establishing a predictive ability independently. Furthermore, this risk score model was compared among identified signatures in previous studies and applied to two external cohorts, showing better prediction performance and generalization ability, and also validated as robust in association with immune cell infiltration in bulk tissues. Besides, a transcription factor regulation network suggested upper regulatory mechanisms in OV. Our immune risk score model may provide gyneco-oncologists with predictive values for the prognosis and treatment management of patients with OV.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guangmei Zhang
- Department of Gynecology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Altamirano GA, Gomez AL, Schierano-Marotti G, Muñoz-de-Toro M, Rodriguez HA, Kass L. Bisphenol A and benzophenone-3 exposure alters milk protein expression and its transcriptional regulation during functional differentiation of the mammary gland in vitro. ENVIRONMENTAL RESEARCH 2020; 191:110185. [PMID: 32946892 DOI: 10.1016/j.envres.2020.110185] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/03/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
The plastic monomer and plasticizer bisphenol A (BPA), and the UV-filter benzophenone-3 (BP3) have been shown to have estrogenic activities that could alter mammary gland development. Our aim was to analyze whether BPA or BP3 direct exposure affects the functional differentiation of the mammary gland using an in vitro model. Mammary organoids were obtained and isolated from 8 week-old virgin female C57BL/6 mice and were differentiated on Matrigel with medium containing lactogenic hormones and exposed to: a) vehicle (0.01% ethanol); b) 1 × 10-9 M or 1 × 10-6 M BPA; or c) 1 × 10-12 M, 1 × 10-9 M or 1 × 10-6 M BP3 for 72 h. The mRNA and protein expression of estrogen receptor alpha (ESR1) and progesterone receptor (PR) were assessed. In addition, mRNA levels of PR-B isoform, glucocorticoid receptor (GR), prolactin receptor (PRLR) and Stat5a, and protein expression of pStat5a/b were evaluated at 72 h. The mRNA and protein expression of milk proteins and their DNA methylation status were also analyzed. Although mRNA level of PRLR and GR was similar between treatments, mRNA expression of ESR1, total PR, PR-B and Stat5a was increased in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. Total PR expression was also increased with 1 × 10-6 M BPA. Nuclear ESR1 and PR expression was observed in all treated organoids; whereas nuclear pStat5a/b alveolar cells was observed only in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. The beta-casein mRNA level was increased in both BPA concentrations and 1 × 10-12 M BP3, which was associated with hypomethylation of its promoter. The beta-casein protein expression was only increased with 1 × 10-9 M BPA or 1 × 10-12 M BP3. In contrast, BPA exposure decreased alpha-lactalbumin mRNA expression and increased DNA methylation level in different methylation-sensitive sites of the gene. Also, 1 × 10-9 M BPA decreased alpha-lactalbumin protein expression. Our results demonstrate that BPA or BP3 exposure alters milk protein synthesis and its transcriptional regulation during mammary gland differentiation in vitro.
Collapse
Affiliation(s)
- Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ayelen L Gomez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gonzalo Schierano-Marotti
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Horacio A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
22
|
The not-so-sweet side of sugar: Influence of the microenvironment on the processes that unleash cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165960. [PMID: 32919034 DOI: 10.1016/j.bbadis.2020.165960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022]
Abstract
The role of "aerobic glycolysis" in cancer has been examined often in the past. Results from those studies, most of which were performed on two dimensional conditions (2D, tissue culture plastic), demonstrate that aerobic glycolysis occurs as a consequence of oncogenic events. These oncogenic events often drive malignant cell growth and survival. Although 2D based experiments are useful in elucidating the molecular mechanisms of oncogenesis, they fail to take contributions of the extracellular microenvironment into account. Indeed we, and others, have shown that the cellular microenvironment is essential in regulating processes that induce and/or suppress the malignant phenotype/properties. This regulation between the cell and its microenvironment is both dynamic and reciprocal and involves the integration of cellular signaling networks in the right context. Therefore, given our previous demonstration of the effect of the microenvironment including tissue architecture and media composition on gene expression and the integration of signaling events observed in three-dimension (3D), we hypothesized that glucose uptake and metabolism must also be essential components of the tissue's signal "integration plan" - that is, if uptake and metabolism of glucose were hyperactivated, the canonical oncogenic pathways should also be similarly activated. This hypothesis, if proven true, suggests that direct inhibition of glucose metabolism in cancer cells should either suppress or revert the malignant phenotype in 3D. Here, we review the up-to-date progress that has been made towards understanding the role that glucose metabolism plays in oncogenesis and re-establishing basally polarized acini in malignant human breast cells.
Collapse
|
23
|
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: 27] [Impact Index Per Article: 5.4] [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
|
24
|
Padmanaban V, Grasset EM, Neumann NM, Fraser AK, Henriet E, Matsui W, Tran PT, Cheung KJ, Georgess D, Ewald AJ. Organotypic culture assays for murine and human primary and metastatic-site tumors. Nat Protoc 2020; 15:2413-2442. [PMID: 32690957 PMCID: PMC8202162 DOI: 10.1038/s41596-020-0335-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 04/16/2020] [Indexed: 01/20/2023]
Abstract
Cancer invasion and metastasis are challenging to study in vivo since they occur deep inside the body over extended time periods. Organotypic 3D culture of fresh tumor tissue enables convenient real-time imaging, genetic and microenvironmental manipulation and molecular analysis. Here, we provide detailed protocols to isolate and culture heterogenous organoids from murine and human primary and metastatic site tumors. The time required to isolate organoids can vary based on the tissue and organ type but typically takes <7 h. We describe a suite of assays that model specific aspects of metastasis, including proliferation, survival, invasion, dissemination and colony formation. We also specify comprehensive protocols for downstream applications of organotypic cultures that will allow users to (i) test the role of specific genes in regulating various cellular processes, (ii) distinguish the contributions of several microenvironmental factors and (iii) test the effects of novel therapeutics.
Collapse
Affiliation(s)
- Veena Padmanaban
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eloise M. Grasset
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Neil M. Neumann
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Andrew K. Fraser
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Elodie Henriet
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - William Matsui
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Phuoc T. Tran
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kevin J. Cheung
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dan Georgess
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Beirut, Lebanon
| | - Andrew J. Ewald
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Author for Correspondence: Andrew J. Ewald, 855 N. Wolfe Street, Rangos 452, Baltimore, MD 21205, Tel: 410-614-9288,
| |
Collapse
|
25
|
Beksaç B, İlter N, Erdem Ö, Çakmak P, Çenetoğlu S, Yapar D. Sparsity of dendritic cells and cytotoxic T cells in tumor microenvironment may lead to recurrence in basal cell carcinoma. Int J Dermatol 2020; 59:1258-1263. [PMID: 32686125 DOI: 10.1111/ijd.15065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/30/2020] [Accepted: 06/18/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Antitumor immune response affects tumor growth. The effect of antitumor immune response on recurrence has been poorly studied in basal cell carcinoma (BCC). OBJECTIVES To investigate the effects of the peritumoral immune infiltrate on BCC recurrence. METHODS A total of 30 BCC patients without recurrence and 29 BCC patients with recurrence were included in this retrospective study. Non-recurrent tumor samples as well as primary and recurrent tumor samples from the recurrent group were stained immunohistochemically with anti-CD4, CD8, CD25, FOXP3, CD68, CD163, and CD1a antibodies. Immune infiltrates were semiquantitatively evaluated. RESULTS BCC tumor microenvironment was rich in CD4+ cells. CD163 expression was higher than CD68. In primary tumors of the recurrent group, CD8 expression was significantly lower than CD4 expression. CD1a expression was lower in primary tumors of the recurrent group than in nonrecurrent tumors. CONCLUSIONS Our results suggest the existence of an immunosuppressive microenvironment in BCC. Lower CD8+ T-cell numbers and sparsity of dendritic cells in primary tumors of recurrent patients suggest further immunosuppression in the tumor microenvironment and an increase in recurrence risk. This is the first study that evaluates and compares tumor immune microenvironments of primary and recurrent BCC lesions with several markers and investigates the role of antitumor immunity on BCC recurrence.
Collapse
Affiliation(s)
- Burcu Beksaç
- Department of Dermatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Nilsel İlter
- Department of Dermatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Özlem Erdem
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Pinar Çakmak
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Seyhan Çenetoğlu
- Department of Plastic, Reconstructive and Aesthetic Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Dilek Yapar
- Department of Public Health, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
26
|
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
|
27
|
Da Silva L, Bray JK, Bulut G, Jiang J, Schmittgen TD. Method for improved integrity of RNA isolated from Matrigel cultures. MethodsX 2020; 7:100966. [PMID: 32637337 PMCID: PMC7327238 DOI: 10.1016/j.mex.2020.100966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/12/2020] [Indexed: 12/28/2022] Open
Abstract
Matrigel is a commercially available substrate that is derived from the extracellular matrix. Matrigel is widely used in cell culture experiments such as the transdifferentiation of primary pancreatic acini to ductal epithelial-like cells. Difficulty arises during gene expression analysis for cells cultured on Matrigel because residual RNA in the Matrigel will not only contribute to the poor integrity of RNA isolated from Matrigel cultures, but also will impact the gene expression data. We report here a simple method of removing Matrigel from primary cultures of human or mouse pancreatic acini. Following the experiment, the cultures are placed on wet ice to liquefy the Matrigel. The cell and Matrigel mixture is then centrifuged at low speed to separate the pancreatic cells from the Matrigel solution that resides in the supernatant. RNA isolated from the pelleted cells has high integrity and may be readily used for gene expression analysis such as quantitative reverse transcription PCR.
Collapse
Affiliation(s)
- Lais Da Silva
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Julie K. Bray
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Gamze Bulut
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Jinmai Jiang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL USA
| | - Thomas D. Schmittgen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL USA
| |
Collapse
|
28
|
Yang N, Xiao W, Song X, Wang W, Dong X. Recent Advances in Tumor Microenvironment Hydrogen Peroxide-Responsive Materials for Cancer Photodynamic Therapy. NANO-MICRO LETTERS 2020; 12:15. [PMID: 34138092 PMCID: PMC7770924 DOI: 10.1007/s40820-019-0347-0] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/17/2019] [Indexed: 05/21/2023]
Abstract
Photodynamic therapy (PDT), as one of the noninvasive clinical cancer phototherapies, suffers from the key drawback associated with hypoxia at the tumor microenvironment (TME), which plays an important role in protecting tumor cells from damage caused by common treatments. High concentration of hydrogen peroxide (H2O2), one of the hallmarks of TME, has been recognized as a double-edged sword, posing both challenges, and opportunities for cancer therapy. The promising perspectives, strategies, and approaches for enhanced tumor therapies, including PDT, have been developed based on the fast advances in H2O2-enabled theranostic nanomedicine. In this review, we outline the latest advances in H2O2-responsive materials, including organic and inorganic materials for enhanced PDT. Finally, the challenges and opportunities for further research on H2O2-responsive anticancer agents are envisioned .
Collapse
Affiliation(s)
- Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China
| | - Wanyue Xiao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China.
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng, 252059, People's Republic of China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China.
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing, 210044, People's Republic of China.
| |
Collapse
|
29
|
Yang N, Xiao W, Song X, Wang W, Dong X. Recent Advances in Tumor Microenvironment Hydrogen Peroxide-Responsive Materials for Cancer Photodynamic Therapy. NANO-MICRO LETTERS 2020; 12:15. [PMID: 34138092 DOI: 10.3847/1538-4357/ab5f08] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/17/2019] [Indexed: 05/27/2023]
Abstract
Photodynamic therapy (PDT), as one of the noninvasive clinical cancer phototherapies, suffers from the key drawback associated with hypoxia at the tumor microenvironment (TME), which plays an important role in protecting tumor cells from damage caused by common treatments. High concentration of hydrogen peroxide (H2O2), one of the hallmarks of TME, has been recognized as a double-edged sword, posing both challenges, and opportunities for cancer therapy. The promising perspectives, strategies, and approaches for enhanced tumor therapies, including PDT, have been developed based on the fast advances in H2O2-enabled theranostic nanomedicine. In this review, we outline the latest advances in H2O2-responsive materials, including organic and inorganic materials for enhanced PDT. Finally, the challenges and opportunities for further research on H2O2-responsive anticancer agents are envisioned .
Collapse
Affiliation(s)
- Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China
| | - Wanyue Xiao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China.
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng, 252059, People's Republic of China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211800, People's Republic of China.
- School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing, 210044, People's Republic of China.
| |
Collapse
|
30
|
COX-2 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1277:87-104. [PMID: 33119867 DOI: 10.1007/978-3-030-50224-9_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumorigenesis is a multistep, complicated process, and many studies have been completed over the last few decades to elucidate this process. Increasingly, many studies have shifted focus toward the critical role of the tumor microenvironment (TME), which consists of cellular players, cell-cell communications, and extracellular matrix (ECM). In the TME, cyclooxygenase-2 (COX-2) has been found to be a key molecule mediating the microenvironment changes. COX-2 is an inducible form of the enzyme that converts arachidonic acid into the signal transduction molecules (thromboxanes and prostaglandins). COX-2 is frequently expressed in many types of cancers and has been closely linked to its occurrence, progression, and prognosis. For example, COX-2 has been shown to (1) regulate tumor cell growth, (2) promote tissue invasion and metastasis, (3) inhibit apoptosis, (4) suppress antitumor immunity, and (5) promote sustainable angiogenesis. In this chapter, we summarize recent advances of studies that have evaluated COX-2 signaling in TME.
Collapse
|
31
|
Fifield BA, Qemo I, Kirou E, Cardiff RD, Porter LA. The atypical cyclin-like protein Spy1 overrides p53-mediated tumour suppression and promotes susceptibility to breast tumourigenesis. Breast Cancer Res 2019; 21:140. [PMID: 31829284 PMCID: PMC6907270 DOI: 10.1186/s13058-019-1211-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 10/15/2019] [Indexed: 01/09/2023] Open
Abstract
Background Breast cancer is the most common cancer to affect women and one of the leading causes of cancer-related deaths. Proper regulation of cell cycle checkpoints plays a critical role in preventing the accumulation of deleterious mutations. Perturbations in the expression or activity of mediators of cell cycle progression or checkpoint activation represent important events that may increase susceptibility to the onset of carcinogenesis. The atypical cyclin-like protein Spy1 was isolated in a screen for novel genes that could bypass the DNA damage response. Clinical data demonstrates that protein levels of Spy1 are significantly elevated in ductal and lobular carcinoma of the breast. We hypothesized that elevated Spy1 would override protective cell cycle checkpoints and support the onset of mammary tumourigenesis. Methods We generated a transgenic mouse model driving expression of Spy1 in the mammary epithelium. Mammary development, growth characteristics and susceptibility to tumourigenesis were studied. In vitro studies were conducted to investigate the relationship between Spy1 and p53. Results We found that in the presence of wild-type p53, Spy1 protein is held ‘in check’ via protein degradation, representing a novel endogenous mechanism to ensure protected checkpoint control. Regulation of Spy1 by p53 is at the protein level and is mediated in part by Nedd4. Mutation or abrogation of p53 is sufficient to allow for accumulation of Spy1 levels resulting in mammary hyperplasia. Sustained elevation of Spy1 results in elevated proliferation of the mammary gland and susceptibility to tumourigenesis. Conclusions This mouse model demonstrates for the first time that degradation of the cyclin-like protein Spy1 is an essential component of p53-mediated tumour suppression. Targeting cyclin-like protein activity may therefore represent a mechanism of re-sensitizing cells to important cell cycle checkpoints in a therapeutic setting.
Collapse
Affiliation(s)
- Bre-Anne Fifield
- Department of Biological Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Ingrid Qemo
- Department of Biological Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Evie Kirou
- Department of Biological Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Robert D Cardiff
- Center of Comparative Medicine, University of California, Davis, CA, USA
| | - Lisa Ann Porter
- Department of Biological Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
32
|
Pemathilaka RL, Reynolds DE, Hashemi NN. Drug transport across the human placenta: review of placenta-on-a-chip and previous approaches. Interface Focus 2019; 9:20190031. [PMID: 31485316 PMCID: PMC6710654 DOI: 10.1098/rsfs.2019.0031] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/20/2022] Open
Abstract
In the past few decades, the placenta became a very controversial topic that has had many researchers and pharmacists discussing the significance of the effects of pharmaceutical drug intake and how it is a possible leading cause towards birth defects. The creation of an in vitro microengineered model of the placenta can be used to replicate the interactions between the mother and fetus, specifically pharmaceutical drug intake reactions. As the field of nanotechnology significantly continues growing, nanotechnology will become more apparent in the study of medicine and other scientific disciplines, specifically microengineering applications. This review is based on past and current research that compares the feasibility and testing of the placenta-on-a-chip microengineered model to the previous and underdeveloped in vivo and ex vivo approaches. The testing of the practicality and effectiveness of the in vitro, in vivo and ex vivo models requires the experimentation of prominent pharmaceutical drugs that most mothers consume during pregnancy. In this case, these drugs need to be studied and tested more often. However, there are challenges associated with the in vitro, in vivo and ex vivo processes when developing a practical placental model, which are discussed in further detail.
Collapse
Affiliation(s)
| | - David E. Reynolds
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
| | - Nicole N. Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
33
|
CD155 expression in human breast cancer: Clinical significance and relevance to natural killer cell infiltration. Life Sci 2019; 231:116543. [DOI: 10.1016/j.lfs.2019.116543] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
|
34
|
Ma G, Gao A, Yang Y, He Y, Zhang X, Zhang B, Zhang Z, Li M, Fu X, Zhao D, Wu R, Qi L, Hu Q, Li J, Fu L, Zhu Z, Dong JT. Zfhx3 is essential for progesterone/progesterone receptor signaling to drive ductal side-branching and alveologenesis in mouse mammary glands. J Genet Genomics 2019; 46:119-131. [PMID: 30954439 DOI: 10.1016/j.jgg.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/05/2019] [Indexed: 01/26/2023]
Abstract
Progesterone (Pg)/progesterone receptor (PR) signaling drives mammary gland side-branching and alveologenesis, but the mechanisms through which Pg/PR signaling functions remain to be clarified. Using in vitro and in vivo models and histological and molecular analyses, we determined the role of Zfhx3 transcription factor in mammary gland development driven by Pg/PR signaling. Postnatal deletion of Zfhx3 in mouse mammary epithelial cells attenuated side-branching morphogenesis and alveologenesis. These effects were undetectable in the absence of Pg/PR signaling. During the estrus cycle, Zfhx3 expression corresponded to that of Pg, being at the highest level at the diestrus stage; Zfhx3 deletion inhibited mammary gland branching more potently at diestrus than estrus stage. Loss of Zfhx3 not only attenuated the expansion of stem/progenitor cells driven by Pg/PR signaling, but also impaired the function of Pg/PR signaling in the transcriptional activation of multiple genes. In addition, Pg/PR signaling significantly expanded PR- and Zfhx3-positive epithelial cells, and induced the physical association of ZFHX3 with PR. These findings establish Zfhx3 as an integral transcription factor of Pg/PR signaling in driving side-branching and alveologenesis during mammary gland development.
Collapse
Affiliation(s)
- Gui Ma
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ang Gao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yinan Yang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuan He
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xi Zhang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Baotong Zhang
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Zhiqian Zhang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Mei Li
- Ningbo Institute of Medical Sciences, Ningbo, 315020, China
| | - Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dan Zhao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Leilei Qi
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qingxia Hu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Juan Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China; Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
35
|
Cruz-Acuña R, García AJ. Engineered materials to model human intestinal development and cancer using organoids. Exp Cell Res 2019; 377:109-114. [PMID: 30794801 DOI: 10.1016/j.yexcr.2019.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/27/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023]
Abstract
Human organoids provide constructive in vitro models of human development and disease, as these recapitulate important morphogenetic and functional features of the tissue and species of origin. However, organoid culture technologies often involve the use of biologically-derived materials (e.g. Matrigel™) that do not allow dissection of the independent contributions of the biochemical and biophysical matrix properties to organoid development. Additionally, their inherent lot-to-lot variability and, in the case of Matrigel™, tumor-derived nature limits their applicability as platforms for drug and tissue transplantation therapies. Here, we highlight recent studies that overcome these limitations through engineering of novel biomaterial platforms that (1) allow to study the independent contributions of physicochemical matrix properties to organoid development and their potential for translational therapies, and (2) better recreate the tumor microenvironment for high-throughput, pre-clinical drug development. These studies illustrate how innovative biomaterial constructs can contribute to the modeling of human development and disease using organoids, and as platforms for development of organoid-based therapies. Finally, we discuss the current limitations of the organoid field and how they can potentially be addressed using engineered biomaterials.
Collapse
Affiliation(s)
- Ricardo Cruz-Acuña
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States; Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| |
Collapse
|
36
|
Søgaard PP, Ito N, Sato N, Fujita Y, Matter K, Itoh Y. Epithelial polarization in 3D matrix requires DDR1 signaling to regulate actomyosin contractility. Life Sci Alliance 2019; 2:2/1/e201800276. [PMID: 30760555 PMCID: PMC6374992 DOI: 10.26508/lsa.201800276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 01/19/2023] Open
Abstract
For epithelial cells to establish epithelial polarity in a 3D matrix, signaling of a collagen receptor tyrosine kinase, DDR1, plays a crucial role. DDR1 signaling controls actomyosin contractility at the cell–cell junction through suppression of ROCK activity. Epithelial cells form sheets and tubules in various epithelial organs and establish apicobasal polarity and asymmetric vesicle transport to provide functionality in these structures. However, the molecular mechanisms that allow epithelial cells to establish polarity are not clearly understood. Here, we present evidence that the kinase activity of the receptor tyrosine kinase for collagen, discoidin domain receptor 1 (DDR1), is required for efficient establishment of epithelial polarity, proper asymmetric protein secretion, and execution of morphogenic programs. Lack of DDR1 protein or inhibition of DDR1 kinase activity disturbed tubulogenesis, cystogenesis, and the establishment of epithelial polarity and caused defects in the polarized localization of membrane-type 1 matrix metalloproteinase (MT1-MMP), GP135, primary cilia, laminin, and the Golgi apparatus. Disturbed epithelial polarity and cystogenesis upon DDR1 inhibition was caused by excess ROCK (rho-associated, coiled-coil-containing protein kinase)-driven actomyosin contractility, and pharmacological inhibition of ROCK was sufficient to correct these defects. Our data indicate that a DDR1-ROCK signaling axis is essential for the efficient establishment of epithelial polarity.
Collapse
Affiliation(s)
| | - Noriko Ito
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Nanami Sato
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Yasuyuki Fujita
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Karl Matter
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
37
|
Jogalekar MP, Serrano EE. Total RNA Isolation from Separately Established Monolayer and Hydrogel Cultures of Human Glioblastoma Cell Line. Bio Protoc 2019; 9:e3305. [PMID: 31528666 DOI: 10.21769/bioprotoc.3305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Astrocytoma is an invasive carcinoma occurring in the nervous system and currently lacks effective treatment options. A deeper understanding of the mechanisms of tumorigenesis and tumor progression is needed in order to develop novel therapeutic strategies. Recent advances in in vitro culture systems have demonstrated that the use of three-dimensional (3D) culture models could be more relevant for this purpose as compared to monolayer or two-dimensional (2D) models due to their resemblance to in vivo cancer pathology. High-throughput techniques such as RNA sequencing, microarray analyses and cloning could provide useful insights into the relevance of these systems to the native tissue. Previous studies have reported RNA extraction protocols needed for such applications. We have modified these protocols to suit the isolation of total RNA from monolayer and hydrogel cultures of astrocytoma established using basement membrane matrix, Geltrex™. We have used this method to demonstrate the differences in the expression of genes involved in autophagy, a process deregulated in many cancer types, in monolayer and hydrogel cultures using quantitative polymerase chain reaction (qPCR). This protocol can be adopted by the researchers who wish to understand the molecular basis of gene expression in hydrogel cultures of normal as well as cancer cell lines.
Collapse
Affiliation(s)
- Manasi P Jogalekar
- Molecular Biology Program, New Mexico State University, Las Cruces, NM, USA
| | - Elba E Serrano
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
38
|
Zhang H, Fredericks T, Xiong G, Qi Y, Rychahou PG, Li JD, Pihlajaniemi T, Xu W, Xu R. Membrane associated collagen XIII promotes cancer metastasis and enhances anoikis resistance. Breast Cancer Res 2018; 20:116. [PMID: 30285809 PMCID: PMC6167877 DOI: 10.1186/s13058-018-1030-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Increased collagen expression and deposition are associated with cancer progression and poor prognosis in breast cancer patients. However, function and regulation of membrane-associated collagen in breast cancer have not been determined. Collagen XIII is a type II transmembrane protein within the collagen superfamily. Experiments in tissue culture and knockout mouse models show that collagen XIII is involved in cell adhesion and differentiation of certain cell types. In the present study, we determined roles of collagen XIII in breast cancer progression and metastasis. METHODS We analyzed the association of collagen XIII expression with breast cancer development and metastasis using published gene expression profiles generated from human breast cancer tissues. Utilizing gain- and loss- of function approaches and 3D culture assays, we investigated roles of collagen XIII in regulating invasive tumor growth. Using the tumorsphere/mammosphere formation assay and the detachment cell culture assay, we determined whether collagen XIII enhances cancer cell stemness and induces anoikis resistance. We also inhibited collagen XIII signaling with β1 integrin function-blocking antibody. Finally, using the lung colonization assay and the orthotopic mammary tumor model, we investigated roles of collagen XIII in regulating breast cancer colonization and metastasis. Cox proportional hazard (log-rank) test, two-sided Student's t-test (two groups) and one-way ANOVA (three or more groups) analyses were used in this study. RESULTS Collagen XIII expression is significantly higher in human breast cancer tissue compared with normal mammary gland. Increased collagen XIII mRNA levels in breast cancer tissue correlated with short distant recurrence free survival. We showed that collagen XIII expression promoted invasive tumor growth in 3D culture, enhanced cancer cell stemness, and induced anoikis resistance. Collagen XIII expression induced β1 integrin activation. Blocking β1 integrin activation significantly reduced collagen XIII-induced invasion and mammosphere formation. Importantly, silencing collagen XIII in MDA-MB-231 cells reduced lung colonization and metastasis. CONCLUSIONS Our results demonstrate a novel function of collagen XIII in promoting cancer metastasis, cell invasion, and anoikis resistance.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.,UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Tricia Fredericks
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY, 40504, USA
| | - Gaofeng Xiong
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Yifei Qi
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Piotr G Rychahou
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY, 40504, USA
| | - Jia-Da Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan Province, China
| | - Taina Pihlajaniemi
- Center for Cell-Matrix Research and Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014, Oulu, Finland
| | - Wei Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| | - Ren Xu
- UK Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA. .,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
39
|
Amara N, Tholen M, Bogyo M. Chemical Tools for Selective Activity Profiling of Endogenously Expressed MMP-14 in Multicellular Models. ACS Chem Biol 2018; 13:2645-2654. [PMID: 30160940 DOI: 10.1021/acschembio.8b00562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Matrix metalloproteases (MMPs) are a large family of zinc-dependent endopeptidases involved in a diverse set of physiological and pathological processes, most notably in cancer. Current methods for imaging and quantifying MMP activity lack sufficient selectivity and spatiotemporal resolution to allow studies of specific MMP function in vivo. Previously, we reported a strategy for selective targeting of MMPs by engineering a functionally silent cysteine mutation that enables highly specific covalent modification by a designed activity-based probe. Here, we describe the translation of that technology into a mouse model of breast cancer and subsequent demonstration of the utility of the approach for studies of MMP-14 activation in the tumor microenvironment. Using this approach, we find that MMP-14 is active in late stage tumors and is predominantly associated with stromal cell populations that have been activated by specific signaling molecules (e.g., TGFβ) produced by tumor cells. Our data demonstrate the applicability of this approach for studies of MMP function in whole organisms and identify important regulatory mechanisms for MMP-14 activity in the tumor microenvironment.
Collapse
|
40
|
Kaushik G, Ponnusamy MP, Batra SK. Concise Review: Current Status of Three-Dimensional Organoids as Preclinical Models. Stem Cells 2018; 36:1329-1340. [PMID: 29770526 DOI: 10.1002/stem.2852] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/10/2018] [Accepted: 05/01/2018] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cultures use the property of some cells to self-organize in matrices and generate structures that can be programmed to represent an organ or a pathology. Organoid cultures are the 3D cultivation of source tissue (ranging from cells to tissue fragments) in a support matrix and specialized media that nearly resembles the physiological environment. Depending on the source tissue, growth factors, and inhibitors provided, organoids can be programmed to recapitulate the biology of a system and progression of pathology. Organoids are genetically stable, and genetically amenable, making them very suitable tools to study tissue homeostasis and cancer. In this Review, we focus on providing recent technical advances from published literature to efficiently use organoids as a tool for disease modeling and therapeutics. Also, we discuss stem cell biology principles used to generate multiple organoids and their characteristics, with a brief description of methodology. A major theme of this review is to expand organoid applications to the study disease progression and drug response in different cancers. We also discuss shortcomings, limitations, and advantages of developed 3D cultures, with the rationale behind the methodology. Stem Cells 2018;36:1329-1340.
Collapse
Affiliation(s)
- Garima Kaushik
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
41
|
Kovac B, Mäkelä TP, Vallenius T. Increased α-actinin-1 destabilizes E-cadherin-based adhesions and associates with poor prognosis in basal-like breast cancer. PLoS One 2018; 13:e0196986. [PMID: 29742177 PMCID: PMC5942811 DOI: 10.1371/journal.pone.0196986] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/24/2018] [Indexed: 01/04/2023] Open
Abstract
The controlled formation and stabilization of E-cadherin-based adhesions is vital for epithelial integrity. This requires co-operation between the E-cadherin-based adhesions and the associated actin cytoskeleton. In cancer, this co-operation often fails, predisposing cells to migration through molecular mechanisms that have only been partially characterized. Here, we demonstrate that the actin filament cross-linker α-actinin-1 is frequently increased in human breast cancer. In mammary epithelial cells, the increased α-actinin-1 levels promote cell migration and induce disorganized acini-like structures in Matrigel. This is accompanied by a major reorganization of the actin cytoskeleton and the associated E-cadherin-based adhesions. Increased expression of α-actinin-1 is particularly noted in basal-like breast cancer cell lines, and in breast cancer patients it associates with poor prognosis in basal-like subtypes. Downregulation of α-actinin-1 in E-cadherin expressing basal-like breast cancer cells demonstrate that α-actinin-1-assembled actin fibers destabilize E-cadherin-based adhesions. Taken together, these results indicate that increased α-actinin-1 expression destabilizes E-cadherin-based adhesions, which is likely to promote the migratory potential of breast cancer cells. Furthermore, our results identify α-actinin-1 as a candidate prognostic biomarker in basal-like breast cancer.
Collapse
Affiliation(s)
- Bianca Kovac
- Research Programs Unit, Faculty of Medicine and Helsinki Institute of Life Science HiLIFE, University of Helsinki, Biomedicum Helsinki, Helsinki, Finland
| | - Tomi P. Mäkelä
- Research Programs Unit, Faculty of Medicine and Helsinki Institute of Life Science HiLIFE, University of Helsinki, Biomedicum Helsinki, Helsinki, Finland
| | - Tea Vallenius
- Research Programs Unit, Faculty of Medicine and Helsinki Institute of Life Science HiLIFE, University of Helsinki, Biomedicum Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
42
|
Park J, Wetzel I, Dréau D, Cho H. 3D Miniaturization of Human Organs for Drug Discovery. Adv Healthc Mater 2018; 7. [PMID: 28885786 DOI: 10.1002/adhm.201700551] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/14/2017] [Indexed: 12/15/2022]
Abstract
"Engineered human organs" hold promises for predicting the effectiveness and accuracy of drug responses while reducing cost, time, and failure rates in clinical trials. Multiorgan human models utilize many aspects of currently available technologies including self-organized spherical 3D human organoids, microfabricated 3D human organ chips, and 3D bioprinted human organ constructs to mimic key structural and functional properties of human organs. They enable precise control of multicellular activities, extracellular matrix (ECM) compositions, spatial distributions of cells, architectural organizations of ECM, and environmental cues. Thus, engineered human organs can provide the microstructures and biological functions of target organs and advantageously substitute multiscaled drug-testing platforms including the current in vitro molecular assays, cell platforms, and in vivo models. This review provides an overview of advanced innovative designs based on the three main technologies used for organ construction leading to single and multiorgan systems useable for drug development. Current technological challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Joseph Park
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Isaac Wetzel
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Didier Dréau
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Hansang Cho
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| |
Collapse
|
43
|
Characterization of primary human mammary epithelial cells isolated and propagated by conditional reprogrammed cell culture. Oncotarget 2017; 9:11503-11514. [PMID: 29545915 PMCID: PMC5837767 DOI: 10.18632/oncotarget.23817] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023] Open
Abstract
Purpose Conditional reprogramming methods allow for the inexhaustible in vitro proliferation of primary epithelial cells from human tissue specimens. This methodology has the potential to enhance the utility of primary cell culture as a model for mammary gland research. However, few studies have systematically characterized this method in generating in vitro normal human mammary epithelial cell models. Results We show that cells derived from fresh normal breast tissues can be propagated and exhibit heterogeneous morphologic features. The cultures are composed of CK18, desmoglein 3, and CK19-positive luminal cells and vimentin, p63, and CK14-positive myoepithelial cells, suggesting the maintenance of in vivo heterogeneity. In addition, the cultures contain subpopulations with different CD49f and EpCAM expression profiles. When grown in 3D conditions, cells self-organize into distinct structures that express either luminal or basal cell markers. Among these structures, CK8-positive cells enclosing a lumen are capable of differentiation into milk-producing cells in the presence of lactogenic stimulus. Furthermore, our short-term cultures retain the expression of ERα, as well as its ability to respond to estrogen stimulation. Materials and Methods We have investigated conditionally reprogrammed normal epithelial cells in terms of cell type heterogeneity, cellular marker expression, and structural arrangement in two-dimensional (2D) and three-dimensional (3D) systems. Conclusions The conditional reprogramming methodology allows generation of a heterogeneous culture from normal human mammary tissue in vitro. We believe that this cell culture model will provide a valuable tool to study mammary cell function and malignant transformation.
Collapse
|
44
|
Li D, Xing Y, Li H, Wang W, Hou X, Gao M. Effect of linoleic acid supplementation on triglyceride content and gene expression in milk fat synthesis in two- and three-dimensional cultured bovine mammary epithelial cells. ITALIAN JOURNAL OF ANIMAL SCIENCE 2017. [DOI: 10.1080/1828051x.2017.1412273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Dabiao Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuanyuan Xing
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Honglei Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Weiyun Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Xianzhi Hou
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Min Gao
- Animal Nutrition Institute, Agriculture and Animal Husbandry Academy of Inner Mongolia, Hohhot, China
| |
Collapse
|
45
|
Njoroge RN, Unno K, Zhao JC, Naseem AF, Anker JF, McGee WA, Nonn L, Abdulkadir SA. Organoids model distinct Vitamin E effects at different stages of prostate cancer evolution. Sci Rep 2017; 7:16285. [PMID: 29176677 PMCID: PMC5701195 DOI: 10.1038/s41598-017-16459-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/13/2017] [Indexed: 11/09/2022] Open
Abstract
Vitamin E increased prostate cancer risk in the Selenium and Vitamin E Cancer Prevention Trial (SELECT) through unknown mechanisms while Selenium showed no efficacy. We determined the effects of the SELECT supplements on benign (primary), premalignant ( RWPE-1) and malignant (LNCaP) prostate epithelial organoids. While the supplements decreased proliferation and induced cell death in cancer organoids, they had no effect on the benign organoids. In contrast, Vitamin E enhanced cell proliferation and survival in the premalignant organoids in a manner that recapitulated the SELECT results. Indeed, while Vitamin E induced a pro-proliferative gene expression signature, Selenium alone or combined with Vitamin E produced an anti-proliferative signature. The premalignant organoids also displayed significant downregulation of glucose transporter and glycolytic gene expression pointing to metabolic alterations. Detached RWPE-1 cells had low ATP levels due to diminished glucose uptake and glycolysis which was rescued by Vitamin E through the activation of fatty acid oxidation (FAO). FAO inhibition abrogated the ATP rescue, diminished survival of the inner matrix detached cells, restoring the normal hollow lumen morphology in Vitamin E treated organoids. Organoid models therefore clarify the paradoxical findings from SELECT and demonstrate that Vitamin E promotes tumorigenesis in the early stages of prostate cancer evolution.
Collapse
Affiliation(s)
- Rose N Njoroge
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL, 60611, USA
| | - Kenji Unno
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL, 60611, USA
| | - Jonathan C Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anum F Naseem
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL, 60611, USA
| | - Jonathan F Anker
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL, 60611, USA
| | - Warren A McGee
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Larisa Nonn
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sarki A Abdulkadir
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
46
|
Voura EB, Montalvo MJ, Dela Roca KT, Fisher JM, Defamie V, Narala SR, Khokha R, Mulligan ME, Evans CA. Planarians as models of cadmium-induced neoplasia provide measurable benchmarks for mechanistic studies. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2017; 142:544-554. [PMID: 28482323 DOI: 10.1016/j.ecoenv.2017.04.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/02/2017] [Accepted: 04/19/2017] [Indexed: 06/07/2023]
Abstract
Bioassays of planarian neoplasia highlight the potential of these organisms as useful standards to assess whether environmental toxins such as cadmium promote tumorigenesis. These studies complement other investigations into the exceptional healing and regeneration of planarians - processes that are driven by a population of active stem cells, or neoblasts, which are likely transformed during planarian tumor growth. Our goal was to determine if planarian tumorigenesis assays are amenable to mechanistic studies of cadmium carcinogenesis. To that end we demonstrate, by examining both counts of cell populations by size, and instances of mitosis, that the activity of the stem cell population can be monitored. We also provide evidence that specific biomodulators can affect the potential of planarian neoplastic growth, in that an inhibitor of metalloproteinases effectively blocked the development of the lesions. From these results, we infer that neoblast activity does respond to cadmium-induced tumor growth, and that metalloproteinases are required for the progression of cancer in the planarian.
Collapse
Affiliation(s)
- Evelyn B Voura
- School of Science, Technology and Health Studies, Morrisville State College, 80 Eaton Street, Morrisville, New York 13408, USA.
| | - Melissa J Montalvo
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Kevin T Dela Roca
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Julia M Fisher
- Colgate University, 13 Oak Drive, Hamilton, New York 13346, USA
| | - Virginie Defamie
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Swami R Narala
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Rama Khokha
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Margaret E Mulligan
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Colleen A Evans
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| |
Collapse
|
47
|
Aziz AUR, Geng C, Fu M, Yu X, Qin K, Liu B. The Role of Microfluidics for Organ on Chip Simulations. Bioengineering (Basel) 2017; 4:E39. [PMID: 28952518 PMCID: PMC5590458 DOI: 10.3390/bioengineering4020039] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022] Open
Abstract
A multichannel three-dimensional chip of a microfluidic cell culture which enables the simulation of organs is called an "organ on a chip" (OC). With the integration of many other technologies, OCs have been mimicking organs, substituting animal models, and diminishing the time and cost of experiments which is better than the preceding conventional in vitro models, which make them imperative tools for finding functional properties, pathological states, and developmental studies of organs. In this review, recent progress regarding microfluidic devices and their applications in cell cultures is discussed to explain the advantages and limitations of these systems. Microfluidics is not a solution but only an approach to create a controlled environment, however, other supporting technologies are needed, depending upon what is intended to be achieved. Microfluidic platforms can be integrated with additional technologies to enhance the organ on chip simulations. Besides, new directions and areas are mentioned for interested researchers in this field, and future challenges regarding the simulation of OCs are also discussed, which will make microfluidics more accurate and beneficial for biological applications.
Collapse
Affiliation(s)
- Aziz Ur Rehman Aziz
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Chunyang Geng
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Mengjie Fu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Xiaohui Yu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Kairong Qin
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Bo Liu
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| |
Collapse
|
48
|
Abstract
3D cell culture is an invaluable tool in developmental, cell, and cancer biology. By mimicking crucial features of in vivo environment, including cell-cell and cell-extracellular matrix interactions, 3D cell culture enables proper structural architecture and differentiated function of normal tissues or tumors in vitro. Thereby 3D cell culture realistically models in vivo tissue conditions and processes, and provides in vivo like responses. Since its early days in the 1970s, 3D cell culture has revealed important insights into mechanisms of tissue homeostasis and cancer, and accelerated translational research in cancer biology and tissue engineering.
Collapse
|
49
|
Jardé T, Lloyd-Lewis B, Thomas M, Kendrick H, Melchor L, Bougaret L, Watson PD, Ewan K, Smalley MJ, Dale TC. Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nat Commun 2016; 7:13207. [PMID: 27782124 PMCID: PMC5095178 DOI: 10.1038/ncomms13207] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
The development of in vitro culture systems quantitatively and qualitatively recapitulating normal breast biology is key to the understanding of mammary gland biology. Current three-dimensional mammary culture systems have not demonstrated concurrent proliferation and functional differentiation ex vivo in any system for longer than 2 weeks. Here, we identify conditions including Neuregulin1 and R-spondin 1, allowing maintenance and expansion of mammary organoids for 2.5 months in culture. The organoids comprise distinct basal and luminal compartments complete with functional steroid receptors and stem/progenitor cells able to reconstitute a complete mammary gland in vivo. Alternative conditions are also described that promote enrichment of basal cells organized into multiple layers surrounding a keratinous core, reminiscent of structures observed in MMTV-Wnt1 tumours. These conditions comprise a unique tool that should further understanding of normal mammary gland development, the molecular mechanism of hormone action and signalling events whose deregulation leads to breast tumourigenesis.
Collapse
Affiliation(s)
- Thierry Jardé
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
- Cancer Program, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria 3800, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Bethan Lloyd-Lewis
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Mairian Thomas
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Howard Kendrick
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Lorenzo Melchor
- Division of Breast Cancer Research, Breast Cancer Now, Institute of Cancer Research, London SW3 6JB, UK
| | - Lauriane Bougaret
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Peter D. Watson
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Kenneth Ewan
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Matthew J. Smalley
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Trevor C. Dale
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
50
|
Lin MC, Chen SY, Tsai HM, He PL, Lin YC, Herschman H, Li HJ. PGE 2 /EP 4 Signaling Controls the Transfer of the Mammary Stem Cell State by Lipid Rafts in Extracellular Vesicles. Stem Cells 2016; 35:425-444. [PMID: 27506158 DOI: 10.1002/stem.2476] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 01/09/2023]
Abstract
Prostaglandin E2 (PGE2 )-initiated signaling contributes to stem cell homeostasis and regeneration. However, it is unclear how PGE2 signaling controls cell stemness. This study identifies a previously unknown mechanism by which PGE2 /prostaglandin E receptor 4 (EP4 ) signaling regulates multiple signaling pathways (e.g., PI3K/Akt signaling, TGFβ signaling, Wnt signaling, EGFR signaling) which maintain the basal mammary stem cell phenotype. A shift of basal mammary epithelial stem cells (MaSCs) from a mesenchymal/stem cell state to a non-basal-MaSC state occurs in response to prostaglandin E receptor 4 (EP4 ) antagonism. EP4 antagonists elicit release of signaling components, by controlling their trafficking into extracellular vesicles/exosomes in a lipid raft/caveolae-dependent manner. Consequently, EP4 antagonism indirectly inactivates, through induced extracellular vesicle/exosome release, pathways required for mammary epithelial stem cell homeostasis, e.g. canonical/noncanonical Wnt, TGFβ and PI3K/Akt pathways. EP4 antagonism causes signaling receptors and signaling components to shift from non-lipid raft fractions to lipid raft fractions, and to then be released in EP4 antagonist-induced extracellular vesicles/exosomes, resulting in the loss of the stem cell state by mammary epithelial stem cells. In contrast, luminal mammary epithelial cells can acquire basal stem cell properties following ingestion of EP4 antagonist-induced stem cell extracellular vesicles/exosomes, and can then form mammary glands. These findings demonstrate that PGE2 /EP4 signaling controls homeostasis of mammary epithelial stem cells through regulating extracellular vesicle/exosome release. Reprogramming of mammary epithelial cells can result from EP4 -mediated stem cell property transfer by extracellular vesicles/exosomes containing caveolae-associated proteins, between mammary basal and luminal epithelial cells. Stem Cells 2017;35:425-444.
Collapse
Affiliation(s)
- Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ho-Min Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Lin He
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yen-Chun Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan.,Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Harvey Herschman
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|