1
|
Jose A, Kulkarni P, Thilakan J, Munisamy M, Malhotra AG, Singh J, Kumar A, Rangnekar VM, Arya N, Rao M. Integration of pan-omics technologies and three-dimensional in vitro tumor models: an approach toward drug discovery and precision medicine. Mol Cancer 2024; 23:50. [PMID: 38461268 PMCID: PMC10924370 DOI: 10.1186/s12943-023-01916-6] [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: 08/05/2023] [Accepted: 12/15/2023] [Indexed: 03/11/2024] Open
Abstract
Despite advancements in treatment protocols, cancer is one of the leading cause of deaths worldwide. Therefore, there is a need to identify newer and personalized therapeutic targets along with screening technologies to combat cancer. With the advent of pan-omics technologies, such as genomics, transcriptomics, proteomics, metabolomics, and lipidomics, the scientific community has witnessed an improved molecular and metabolomic understanding of various diseases, including cancer. In addition, three-dimensional (3-D) disease models have been efficiently utilized for understanding disease pathophysiology and as screening tools in drug discovery. An integrated approach utilizing pan-omics technologies and 3-D in vitro tumor models has led to improved understanding of the intricate network encompassing various signalling pathways and molecular cross-talk in solid tumors. In the present review, we underscore the current trends in omics technologies and highlight their role in understanding genotypic-phenotypic co-relation in cancer with respect to 3-D in vitro tumor models. We further discuss the challenges associated with omics technologies and provide our outlook on the future applications of these technologies in drug discovery and precision medicine for improved management of cancer.
Collapse
Affiliation(s)
- Anmi Jose
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Pallavi Kulkarni
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Jaya Thilakan
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Murali Munisamy
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Anvita Gupta Malhotra
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Jitendra Singh
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India
| | - Vivek M Rangnekar
- Markey Cancer Center and Department of Radiation Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Neha Arya
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, 462020, India.
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
2
|
Rodrigues MT, Michelli APP, Caso GF, de Oliveira PR, Rodrigues-Junior DM, Morale MG, Machado Júnior J, Bortoluci KR, Tamura RE, da Silva TRC, Raminelli C, Chau E, Godin B, Calil-Silveira J, Rubio IGS. Lysicamine Reduces Protein Kinase B (AKT) Activation and Promotes Necrosis in Anaplastic Thyroid Cancer. Pharmaceuticals (Basel) 2023; 16:1687. [PMID: 38139812 PMCID: PMC10748177 DOI: 10.3390/ph16121687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Anaplastic thyroid cancer (ATC) is an aggressive form of thyroid cancer (TC), accounting for 50% of total TC-related deaths. Although therapeutic approaches against TC have improved in recent years, the survival rate remains low, and severe adverse effects are commonly reported. However, unexplored alternatives based on natural compounds, such as lysicamine, an alkaloid found in plants with established cytotoxicity against breast and liver cancers, offer promise. Therefore, this study aimed to explore the antineoplastic effects of lysicamine in papillary TC (BCPAP) and ATC (HTH83 and KTC-2) cells. Lysicamine treatment reduced cell viability, motility, colony formation, and AKT activation while increasing the percentage of necrotic cells. The absence of caspase activity confirmed apoptosis-independent cell death. Necrostatin-1 (NEC-1)-mediated necrosome inhibition reduced lysicamine-induced necrosis in KTC-2, suggesting necroptosis induction via a reactive oxygen species (ROS)-independent mechanism. Additionally, in silico analysis predicted lysicamine target proteins, particularly those related to MAPK and TGF-β signaling. Our study demonstrated lysicamine's potential as an antineoplastic compound in ATC cells with a proposed mechanism related to inhibiting AKT activation and inducing cell death.
Collapse
Affiliation(s)
- Mariana Teixeira Rodrigues
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Structural and Functional Biology Post-Graduate Program, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
| | - Ana Paula Picaro Michelli
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
| | - Gustavo Felisola Caso
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
| | - Paloma Ramos de Oliveira
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
| | - Dorival Mendes Rodrigues-Junior
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, 752 36 Uppsala, Sweden;
| | - Mirian Galliote Morale
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
| | - Joel Machado Júnior
- Biological Science Department, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil;
| | - Karina Ramalho Bortoluci
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil;
| | - Rodrigo Esaki Tamura
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
- Biological Science Department, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil;
- Biology–Chemistry Post-Graduate Program, Institute of Environmental, Chemical and Pharmaceutical Science, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil
| | - Tamiris Reissa Cipriano da Silva
- Department of Chemistry, Institute of Environmental, Chemical and Pharmaceutical Science, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil; (T.R.C.d.S.); (C.R.)
| | - Cristiano Raminelli
- Department of Chemistry, Institute of Environmental, Chemical and Pharmaceutical Science, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil; (T.R.C.d.S.); (C.R.)
| | - Eric Chau
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.C.); (B.G.)
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.C.); (B.G.)
- Department of Obstetrics and Gynecology, Weill Cornell Medicine College, New York, NY 10065, USA
| | - Jamile Calil-Silveira
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Health Board III, Universidade Nove de Julho, São Paulo 01525-000, Brazil
| | - Ileana G. Sanchez Rubio
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.T.R.); (A.P.P.M.); (G.F.C.); (P.R.d.O.); (J.C.-S.)
- Structural and Functional Biology Post-Graduate Program, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil
- Cancer Molecular Biology Laboratory, Universidade Federal de São Paulo—UNIFESP, São Paulo 04021-001, Brazil; (M.G.M.); (R.E.T.)
- Biological Science Department, Universidade Federal de São Paulo—UNIFESP, Diadema 09920-000, Brazil;
| |
Collapse
|
3
|
Martínez-Martínez E, Atzei P, Vionnet C, Roubaty C, Kaeser-Pebernard S, Naef R, Dengjel J. A Dual-Acting Nitric Oxide Donor and Phosphodiesterase 5 Inhibitor Activates Autophagy in Primary Skin Fibroblasts. Int J Mol Sci 2022; 23:ijms23126860. [PMID: 35743299 PMCID: PMC9224465 DOI: 10.3390/ijms23126860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Wound healing pathologies are an increasing problem in ageing societies. Chronic, non-healing wounds, which cause high morbidity and severely reduce the quality of life of affected individuals, are frequently observed in aged individuals and people suffering from diseases affected by the Western lifestyle, such as diabetes. Causal treatments that support proper wound healing are still scarce. Here, we performed expression proteomics to study the effects of the small molecule TOP-N53 on primary human skin fibroblasts and keratinocytes. TOP-N53 is a dual-acting nitric oxide donor and phosphodiesterase-5 inhibitor increasing cGMP levels to support proper wound healing. In contrast to keratinocytes, which did not exhibit global proteome alterations, TOP-N53 had profound effects on the proteome of skin fibroblasts. In fibroblasts, TOP-N53 activated the cytoprotective, lysosomal degradation pathway autophagy and induced the expression of the selective autophagy receptor p62/SQSTM1. Thus, activation of autophagy might in part be responsible for beneficial effects of TOP-N53.
Collapse
Affiliation(s)
- Esther Martínez-Martínez
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Paola Atzei
- Topadur Pharma AG, Grabenstrasse 11A, 8952 Schlieren, Switzerland; (P.A.); (R.N.)
| | - Christine Vionnet
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Carole Roubaty
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Stephanie Kaeser-Pebernard
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Reto Naef
- Topadur Pharma AG, Grabenstrasse 11A, 8952 Schlieren, Switzerland; (P.A.); (R.N.)
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
- Correspondence:
| |
Collapse
|
4
|
Poon S, Ailles LE. Modeling the Role of Cancer-Associated Fibroblasts in Tumor Cell Invasion. Cancers (Basel) 2022; 14:962. [PMID: 35205707 PMCID: PMC8870277 DOI: 10.3390/cancers14040962] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
The major cause of cancer-related deaths can be attributed to the metastatic spread of tumor cells-a dynamic and complex multi-step process beginning with tumor cells acquiring an invasive phenotype to allow them to travel through the blood and lymphatic vessels to ultimately seed at a secondary site. Over the years, various in vitro models have been used to characterize specific steps in the cascade to collectively begin providing a clearer picture of the puzzle of metastasis. With the discovery of the TME's supporting role in activating tumor cell invasion and metastasis, these models have evolved in parallel to accommodate features of the TME and to observe its interactions with tumor cells. In particular, CAFs that reside in reactive tumor stroma have been shown to play a substantial pro-invasive role through their matrix-modifying functions; accordingly, this warranted further investigation with the development and use of invasion assays that could include these stromal cells. This review explores the growing toolbox of assays used to study tumor cell invasion, from the simple beginnings of a tumor cell and extracellular matrix set-up to the advent of models that aim to more closely recapitulate the interplay between tumor cells, CAFs and the extracellular matrix. These models will prove to be invaluable tools to help tease out the intricacies of tumor cell invasion.
Collapse
Affiliation(s)
- Stephanie Poon
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Laurie E. Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
5
|
Sari B, Isik M, Eylem CC, Kilic C, Okesola BO, Karakaya E, Emregul E, Nemutlu E, Derkus B. Omics Technologies for High-Throughput-Screening of Cell-Biomaterial Interactions. Mol Omics 2022; 18:591-615. [DOI: 10.1039/d2mo00060a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent research effort in biomaterial development has largely focused on engineering bio-instructive materials to stimulate specific cell signaling. Assessing the biological performance of these materials using time-consuming and trial-and-error traditional...
Collapse
|
6
|
Beller NC, Lukowski JK, Ludwig KR, Hummon AB. Spatial Stable Isotopic Labeling by Amino Acids in Cell Culture: Pulse-Chase Labeling of Three-Dimensional Multicellular Spheroids for Global Proteome Analysis. Anal Chem 2021; 93:15990-15999. [PMID: 34813286 DOI: 10.1021/acs.analchem.1c03461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Three-dimensional cell cultures, or spheroids, are important model systems for cancer research because they recapitulate chemical and phenotypic aspects of in vivo tumors. Spheroids develop radially symmetric chemical gradients, resulting in distinct cellular populations. Stable isotopic labeling by amino acids in cell culture (SILAC) is a well-established approach to quantify protein expression and has previously been used in a pulse-chase format to evaluate temporal changes. In this article, we demonstrate that distinct isotopic signatures can be introduced into discrete spatial cellular populations, effectively tracking proteins to original locations in the spheroid, using a platform that we refer to as spatial SILAC. Spheroid populations were grown with light, medium, and heavy isotopic media, and the concentric shells of cells were harvested by serial trypsinization. Proteins were quantitatively analyzed by ultraperformance liquid chromatography-tandem mass spectrometry. The isotopic signatures correlated with the spatial location and the isotope position do not significantly impact the proteome of each individual layer. Spatial SILAC can be used to examine the proteomic changes in the different layers of the spheroid and to identify protein biomarkers throughout the structure. We show that SILAC labels can be discretely pulsed to discrete positions, without altering the spheroid's proteome, promising future combined pharmacodynamic and pharmacokinetic studies.
Collapse
Affiliation(s)
- Nicole C Beller
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jessica K Lukowski
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Katelyn R Ludwig
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
7
|
Wang M, Yu H, Zhang T, Cao L, Du Y, Xie Y, Ji J, Wu J. In-Depth Comparison of Matrigel Dissolving Methods on Proteomic Profiling of Organoids. Mol Cell Proteomics 2021; 21:100181. [PMID: 34871808 PMCID: PMC8733271 DOI: 10.1016/j.mcpro.2021.100181] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/25/2021] [Accepted: 12/01/2021] [Indexed: 01/06/2023] Open
Abstract
Patient-derived organoids recently emerged as promising ex vivo 3D culture models recapitulating histological and molecular characteristics of original tissues, thus proteomic profiling of organoids could be valuable for function investigation and clinical translation. However, organoids are usually cultured in murine Matrigel (served as scaffolds and matrix), which brings an issue to separate organoids from Matrigel. Because of the complex compositions of Matrigel and thousands of identical peptides shared between Matrigel and organoids, insufficiently dissolved Matrigel could influence proteomic analysis of organoids in multiple ways. Thus, how to dissolve Matrigel matrix and recovery organoid cells efficiently is vital for sample preparation. Here, we comprehensively compared three popular Matrigel dissolving methods (cell recovery solution, dispase, and PBS–EDTA buffer) and investigated the effect of undissolved Matrigel proteins on proteomic profiles of organoids. By integrative analysis of label-free proteomes of Matrigel and stable isotope labeling by amino acids in cell culture proteomes of organoids collected by three methods, respectively, we found that dispase showed an optimal efficiency, with the highest peptide yield and the highest incorporation ratio of stable isotope labeling by amino acids in cell culture labels (97.1%), as well as with the least potential Matrigel contaminants. To help analysis of proteomic profiles of organoids collected by the other two methods, we identified 312 high-confidence Matrigel contaminants, which could be filtered out to attenuate Matrigel interference with minimal loss of biological information. Together, our study identifies bioinformatics and experimental approaches to eliminate interference of Matrigel contaminants efficiently, which will be valuable for basic and translational proteomic research using organoid models. A comprehensive comparison of Matrigel dissolving methods on organoid proteomics. Matrigel leftover influences protein identification and quantification for organoids. Dispase is a satisfying method for proteomic sample preparation of organoids. Exclusion of high-confidence Matrigel contaminants attenuates Matrigel interference.
Collapse
Affiliation(s)
- Man Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Huan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lihua Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuhao Xie
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianmin Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital and Institute, Beijing, China; Peking University International Cancer Institute, Peking University, Beijing, China.
| |
Collapse
|
8
|
Atat OE, Farzaneh Z, Pourhamzeh M, Taki F, Abi-Habib R, Vosough M, El-Sibai M. 3D modeling in cancer studies. Hum Cell 2021; 35:23-36. [PMID: 34761350 DOI: 10.1007/s13577-021-00642-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/31/2021] [Indexed: 01/01/2023]
Abstract
The tumor microenvironment contributes significantly to tumor initiation, progression, and resistance to chemotherapy. Much of our understanding of the tumor and its microenvironment is developed using various methods of cell culture. Throughout the last two decades, research has increasingly shown that 3D cell culture systems can remarkably recapitulate the complexity of tumor architecture and physiology compared to traditional 2D models. Unlike the flat culture system, these novel models enabled more cell-cell and cell-extracellular matrix interactions. By mimicking in vivo microenvironment, 3D culture systems promise to become accurate tools ready to be used in diagnosis, drug screening, and personalized medicine. In this review, we discussed the importance of 3D culture in simulating the tumor microenvironment and focused on the effects of cancer cell-microenvironment interactions on cancer behavior, resistance, proliferation, and metastasis. Finally, we assessed the role of 3D cell culture systems in the contexts of drug screening. 2D culture system is used to study cancer cell growth, progression, behavior, and drug response. It provides contact between cells and supports paracrine crosstalk between host cells and cancer cells. However, this system fails to simulate the architecture and the physiological aspects of in vivo tumor microenvironment due to the absence of cell-cell/ cell-ECM interactions as well as unlimited access to O2 and nutrients, and the absence of tumor heterogeneity. Recently advanced research has led researchers to generate 3D culture system that can better recapitulate the in vivo environment by providing hypoxic medium, facilitating cell-cell and cell-ECM, interactions, and recapitulating heterogeneity of the tumor. Several approaches are used to maintain and expand cancer cells in 3D culture systems such as tumor spheroids (cell aggregate that mimics the in vivo growth of tumor cells), scaffold-based approaches, bioreactors, microfluidic derives, and organoids. 3D systems are currently used for disease modeling and pre-clinical drug testing.
Collapse
Affiliation(s)
- Oula El Atat
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Zahra Farzaneh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatima Taki
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mirvat El-Sibai
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
9
|
Kaczmarczyk JA, Roberts RR, Luke BT, Chan KC, Van Wagoner CM, Felder RA, Saul RG, Simona C, Blonder J. Comparative microsomal proteomics of a model lung cancer cell line NCI-H23 reveals distinct differences between molecular profiles of 3D and 2D cultured cells. Oncotarget 2021; 12:2022-2038. [PMID: 34611477 PMCID: PMC8487723 DOI: 10.18632/oncotarget.28072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the USA and worldwide. Yet, about 95% of new drug candidates validated in preclinical phase eventually fail in clinical trials. Such a high attrition rate is attributed mostly to the inability of conventional two-dimensionally (2D) cultured cancer cells to mimic native three-dimensional (3D) growth of malignant cells in human tumors. To ascertain phenotypical differences between these two distinct culture conditions, we carried out a comparative proteomic analysis of a membrane fraction obtained from 3D- and 2D-cultured NSCLC model cell line NCI-H23. This analysis revealed a map of 1,166 (24%) protein species regulated in culture dependent manner, including differential regulation of a subset of cell surface-based CD molecules. We confirmed exclusive expression of CD99, CD146 and CD239 in 3D culture. Furthermore, label-free quantitation, targeting KRas proteoform-specific peptides, revealed upregulation of both wild type and monoallelic KRas4BG12C mutant at the surface of 3D cultured cells. In order to reduce the high attrition rate of new drug candidates, the results of this study strongly suggests exploiting base-line molecular profiling of a large number of patient-derived NSCLC cell lines grown in 2D and 3D culture, prior to actual drug candidate testing.
Collapse
Affiliation(s)
- Jan A. Kaczmarczyk
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Rhonda R. Roberts
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Brian T. Luke
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - King C. Chan
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Current address: The Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Carly M. Van Wagoner
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Current address: The Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Robin A. Felder
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Richard G. Saul
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Colantonio Simona
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Josip Blonder
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| |
Collapse
|
10
|
Kong JS, Huang X, Choi Y, Yi H, Kang J, Kim S, Kim J, Lee H, Rim YA, Ju JH, Chung WK, Woolf CJ, Jang J, Cho D. Promoting Long-Term Cultivation of Motor Neurons for 3D Neuromuscular Junction Formation of 3D In Vitro Using Central-Nervous-Tissue-Derived Bioink. Adv Healthc Mater 2021; 10:e2100581. [PMID: 34363335 DOI: 10.1002/adhm.202100581] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/16/2021] [Indexed: 12/14/2022]
Abstract
3D cell printing technology is in the spotlight for producing 3D tissue or organ constructs useful for various medical applications. In printing of neuromuscular tissue, a bioink satisfying all the requirements is a challenging issue. Gel integrity and motor neuron activity are two major characters because a harmonious combination of extracellular materials essential to motor neuron activity consists of disadvantages in mechanical properties. Here, a method for fabrication of 3D neuromuscular tissue is presented using a porcine central nervous system tissue decellularized extracellular matrix (CNSdECM) bioink. CNSdECM retains CNS tissue-specific extracellular molecules, provides rheological properties crucial for extrusion-based 3D cell printing, and reveals positive effects on the growth and maturity of axons of motor neurons compared with Matrigel. It also allows long-term cultivation of human-induced-pluripotent-stem-cell-derived lower motor neurons and sufficiently supports their cellular behavior to carry motor signals to muscle fibers. CNSdECM bioink holds great promise for producing a tissue-engineered motor system using 3D cell printing.
Collapse
Affiliation(s)
- Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Xuan Huang
- FM Kirby Neurobiology Center Boston Children's Hospital and Department of Neurobiology Harvard Medical School Boston MA 02115 USA
| | - Yeong‐Jin Choi
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Materials Processing Innovation Research Division Korea Institute of Materials Science (KIMS) 797 10 Changwondaero, Kyungnam Changwon 51508 Republic of Korea
| | - Hee‐Gyeong Yi
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Department of Rural and Biosystems Engineering College of Agriculture and Life Sciences Chonnam National University Gwangju 61186 Republic of Korea
| | - Junsu Kang
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Sejin Kim
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Jongmin Kim
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Hyungseok Lee
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Department of Mechanical and Biomedical Engineering Kangwon National University (KNU) 1 Gangwondaehak‐gil, Seoksa‐dong Chuncheon‐si Gangwon‐do 24341 Republic of Korea
- Interdisciplinary Program in Biohealth‐Machinery Convergence Engineering Kangwon National University (KNU) Chuncheon 24341 Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine The Catholic University of Korea Seoul 137‐701 Republic of Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine The Catholic University of Korea Seoul 137‐701 Republic of Korea
| | - Wan Kyun Chung
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center Boston Children's Hospital and Department of Neurobiology Harvard Medical School Boston MA 02115 USA
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Department of Convergence IT Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology Yonsei University Seoul 03722 Republic of Korea
| | - Dong‐Woo Cho
- School of Interdisciplinary Bioscience and Bioengineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology Yonsei University Seoul 03722 Republic of Korea
| |
Collapse
|
11
|
Scaffold-free 3D cell culture of primary skin fibroblasts induces profound changes of the matrisome. Matrix Biol Plus 2021; 11:100066. [PMID: 34435183 PMCID: PMC8377039 DOI: 10.1016/j.mbplus.2021.100066] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/15/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
The human skin has a highly developed extracellular matrix (ECM) that is vital for proper skin functioning, its 3D architecture playing a pivotal role in support and guidance of resident and invading cells. To establish relevant in vitro models mimicking the complex design observed in vivo, scaffold-based and scaffold-free 3D cell culture systems have been developed. Here we show that scaffold-free systems are well suited for the analysis of ECM protein regulation. Using quantitative mass spectrometry-based proteomics in combination with magnetic 3D bioprinting we characterize changes in the proteome of skin fibroblasts and squamous cell carcinoma cells. Transferring cells from 2D to 3D without any additional scaffold induces a profound upregulation of matrisome proteins indicating the generation of a complex, tissue-like ECM.
Collapse
|
12
|
Schroeder S, Hofer SJ, Zimmermann A, Pechlaner R, Dammbrueck C, Pendl T, Marcello GM, Pogatschnigg V, Bergmann M, Müller M, Gschiel V, Ristic S, Tadic J, Iwata K, Richter G, Farzi A, Üçal M, Schäfer U, Poglitsch M, Royer P, Mekis R, Agreiter M, Tölle RC, Sótonyi P, Willeit J, Mairhofer B, Niederkofler H, Pallhuber I, Rungger G, Tilg H, Defrancesco M, Marksteiner J, Sinner F, Magnes C, Pieber TR, Holzer P, Kroemer G, Carmona-Gutierrez D, Scorrano L, Dengjel J, Madl T, Sedej S, Sigrist SJ, Rácz B, Kiechl S, Eisenberg T, Madeo F. Dietary spermidine improves cognitive function. Cell Rep 2021; 35:108985. [PMID: 33852843 DOI: 10.1016/j.celrep.2021.108985] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 02/08/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Decreased cognitive performance is a hallmark of brain aging, but the underlying mechanisms and potential therapeutic avenues remain poorly understood. Recent studies have revealed health-protective and lifespan-extending effects of dietary spermidine, a natural autophagy-promoting polyamine. Here, we show that dietary spermidine passes the blood-brain barrier in mice and increases hippocampal eIF5A hypusination and mitochondrial function. Spermidine feeding in aged mice affects behavior in homecage environment tasks, improves spatial learning, and increases hippocampal respiratory competence. In a Drosophila aging model, spermidine boosts mitochondrial respiratory capacity, an effect that requires the autophagy regulator Atg7 and the mitophagy mediators Parkin and Pink1. Neuron-specific Pink1 knockdown abolishes spermidine-induced improvement of olfactory associative learning. This suggests that the maintenance of mitochondrial and autophagic function is essential for enhanced cognition by spermidine feeding. Finally, we show large-scale prospective data linking higher dietary spermidine intake with a reduced risk for cognitive impairment in humans.
Collapse
Affiliation(s)
- Sabrina Schroeder
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Raimund Pechlaner
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - G Mark Marcello
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Viktoria Pogatschnigg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Martina Bergmann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Melanie Müller
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Verena Gschiel
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Selena Ristic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Keiko Iwata
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Research Center for Child Mental Development, University of Fukui, 910-1193 Fukui, Japan; Department of Biology, University of Padova, 35121 Padova, Italy
| | - Gesa Richter
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry Medical University of Graz, 8010 Graz, Austria
| | - Aitak Farzi
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Division of Pharmacology, Medical University of Graz (MUG), 8010 Graz, Austria
| | - Muammer Üçal
- Department of Neurosurgery, RU Experimental Neurotraumatology, Medical University Graz, 8036 Graz, Austria
| | - Ute Schäfer
- Department of Neurosurgery, RU Experimental Neurotraumatology, Medical University Graz, 8036 Graz, Austria
| | - Michael Poglitsch
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Philipp Royer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Ronald Mekis
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Marlene Agreiter
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Regine C Tölle
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Péter Sótonyi
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Johann Willeit
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | | | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Defrancesco
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria
| | - Frank Sinner
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria; Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria
| | - Thomas R Pieber
- BioTechMed-Graz, 8010 Graz, Austria; HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria; Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Peter Holzer
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Division of Pharmacology, Medical University of Graz (MUG), 8010 Graz, Austria
| | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Descartes, Université Paris Diderot, Université Sorbonne Paris Cité, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94 805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Suzhou Institute for Systems Biology, Chinese Academy of Sciences, 215123 Suzhou, China; Department of Women's and Children's Health, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | | | - Luca Scorrano
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biology, University of Padova, 35121 Padova, Italy
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Tobias Madl
- BioTechMed-Graz, 8010 Graz, Austria; Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry Medical University of Graz, 8010 Graz, Austria
| | - Simon Sedej
- BioTechMed-Graz, 8010 Graz, Austria; Department of Cardiology, Medical University of Graz, 8036 Graz, Austria; Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Stephan J Sigrist
- Institute of Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; VASCage, Research Centre on Vascular Ageing and Stroke, 6020 Innsbruck, Austria.
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria.
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria.
| |
Collapse
|
13
|
Pieters VM, Co IL, Wu NC, McGuigan AP. Applications of Omics Technologies for Three-Dimensional In Vitro Disease Models. Tissue Eng Part C Methods 2021; 27:183-199. [PMID: 33406987 DOI: 10.1089/ten.tec.2020.0300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Omics technologies, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics, multiomics, and integrated modalities, have greatly contributed to our understanding of various diseases by enabling researchers to probe the molecular wiring of cellular systems in a high-throughput and precise manner. With the development of tissue-engineered three-dimensional (3D) in vitro disease models, such as organoids and spheroids, there is potential of integrating omics technologies with 3D disease models to elucidate the complex links between genotype and phenotype. These 3D disease models have been used to model cancer, infectious disease, toxicity, neurological disorders, and others. In this review, we provide an overview of omics technologies, highlight current and emerging studies, discuss the associated experimental design considerations, barriers and challenges of omics technologies, and provide an outlook on the future applications of omics technologies with 3D models. Overall, this review aims to provide a valuable resource for tissue engineers seeking to leverage omics technologies for diving deeper into biological discovery. Impact statement With the emergence of three-dimensional (3D) in vitro disease models, tissue engineers are increasingly interested to investigate these systems to address biological questions related to disease mechanism, drug target discovery, therapy resistance, and more. Omics technologies are a powerful and high-throughput approach, but their application for 3D disease models is not maximally utilized. This review illustrates the achievements and potential of using omics technologies to leverage the full potential of 3D in vitro disease models. This will improve the quality of such models, advance our understanding of disease, and contribute to therapy development.
Collapse
Affiliation(s)
- Vera M Pieters
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ileana L Co
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Nila C Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
14
|
Migisha Ntwali P, Heo CE, Han JY, Chae SY, Kim M, Vu HM, Kim MS, Kim HI. Mass spectrometry-based proteomics of single cells and organoids: The new generation of cancer research. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.116005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
15
|
Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis 2019; 36:171-198. [PMID: 30972526 DOI: 10.1007/s10585-019-09966-1] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) constitutes the scaffold of tissues and organs. It is a complex network of extracellular proteins, proteoglycans and glycoproteins, which form supramolecular aggregates, such as fibrils and sheet-like networks. In addition to its biochemical composition, including the covalent intermolecular cross-linkages, the ECM is also characterized by its biophysical parameters, such as topography, molecular density, stiffness/rigidity and tension. Taking these biochemical and biophysical parameters into consideration, the ECM is very versatile and undergoes constant remodeling. This review focusses on this remodeling of the ECM under the influence of a primary solid tumor mass. Within this tumor stroma, not only the cancer cells but also the resident fibroblasts, which differentiate into cancer-associated fibroblasts (CAFs), modify the ECM. Growth factors and chemokines, which are tethered to and released from the ECM, as well as metabolic changes of the cells within the tumor bulk, add to the tumor-supporting tumor microenvironment. Metastasizing cancer cells from a primary tumor mass infiltrate into the ECM, which variably may facilitate cancer cell migration or act as barrier, which has to be proteolytically breached by the infiltrating tumor cell. The biochemical and biophysical properties therefore determine the rates and routes of metastatic dissemination. Moreover, primed by soluble factors of the primary tumor, the ECM of distant organs may be remodeled in a way to facilitate the engraftment of metastasizing cancer cells. Such premetastatic niches are responsible for the organotropic preference of certain cancer entities to colonize at certain sites in distant organs and to establish a metastasis. Translational application of our knowledge about the cancer-primed ECM is sparse with respect to therapeutic approaches, whereas tumor-induced ECM alterations such as increased tissue stiffness and desmoplasia, as well as breaching the basement membrane are hallmark of malignancy and diagnostically and histologically harnessed.
Collapse
Affiliation(s)
- Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| |
Collapse
|