1
|
Morgan D, Gardner AL, Brock A. Lineage Tracing Reveals Clone-Specific Responses to Doxorubicin in Triple-Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643980. [PMID: 40166195 PMCID: PMC11956957 DOI: 10.1101/2025.03.18.643980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Triple-negative breast cancer, characterized by aggressive growth and high intratumor heterogeneity, presents a significant clinical challenge. Here, we use a lineage-tracing system, ClonMapper, which couples heritable clonal identifying tags with single-cell RNA-sequencing (scRNA-seq), to better elucidate the response to doxorubicin in a model of TNBC. We demonstrate that, while there is a dose-dependent reduction in overall clonal diversity, there is no pre-existing resistance signature among surviving clones. Separately, we found the existence of two transcriptomically distinct clonal subpopulations that remain through the course of treatment. Among clones persisting across multiple samples we identified divergent phenotypes, suggesting a response to treament independent of clonal identity. Finally, a subset of clones harbor novel changes in expression following treatment. The clone and sample specific responses to treatment identified herein highlight the need for better personalized treatment strategies to overcome tumor heterogeneity.
Collapse
Affiliation(s)
- Daylin Morgan
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, TX, USA
| | - Andrea L. Gardner
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, TX, USA
| | - Amy Brock
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
2
|
Venugopal Menon N, Lee J, Tang T, Lim CT. Microfluidics for morpholomics and spatial omics applications. LAB ON A CHIP 2025; 25:752-763. [PMID: 39865877 DOI: 10.1039/d4lc00869c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Creative designs, precise fluidic manipulation, and automation have supported the development of microfluidics for single-cell applications. Together with the advancements in detection technologies and artificial intelligence (AI), microfluidic-assisted platforms have been increasingly used for new modalities of single-cell investigations and in spatial omics applications. This review explores the use of microfluidic technologies for morpholomics and spatial omics with a focus on single-cell and tissue characterization. We emphasize how various fluid dynamic principles and unique design integrations enable highly precise fluid manipulation, enhancing sample handling in morpholomics. Additionally, we examine the use of microfluidics-assisted spatial barcoding with micrometer resolutions for the spatial profiling of tissue specimens. Finally, we discuss how microfluidics can serve as a bridge for integrating multiple unique fields in omics research and outline key challenges that these technologies may face in practical translation.
Collapse
Affiliation(s)
- Nishanth Venugopal Menon
- Mechanobiology Institute, National University of Singapore, Singapore, 117411 Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| | - Jeeyeon Lee
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599 Singapore
| | - Tao Tang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore, 117411 Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599 Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| |
Collapse
|
3
|
Laisné M, Lupien M, Vallot C. Epigenomic heterogeneity as a source of tumour evolution. Nat Rev Cancer 2025; 25:7-26. [PMID: 39414948 DOI: 10.1038/s41568-024-00757-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
In the past decade, remarkable progress in cancer medicine has been achieved by the development of treatments that target DNA sequence variants. However, a purely genetic approach to treatment selection is hampered by the fact that diverse cell states can emerge from the same genotype. In multicellular organisms, cell-state heterogeneity is driven by epigenetic processes that regulate DNA-based functions such as transcription; disruption of these processes is a hallmark of cancer that enables the emergence of defective cell states. Advances in single-cell technologies have unlocked our ability to quantify the epigenomic heterogeneity of tumours and understand its mechanisms, thereby transforming our appreciation of how epigenomic changes drive cancer evolution. This Review explores the idea that epigenomic heterogeneity and plasticity act as a reservoir of cell states and therefore as a source of tumour evolution. Best practices to quantify epigenomic heterogeneity and explore its various causes and consequences are discussed, including epigenomic reprogramming, stochastic changes and lasting memory. The design of new therapeutic approaches to restrict epigenomic heterogeneity, with the long-term objective of limiting cancer development and progression, is also addressed.
Collapse
Affiliation(s)
- Marthe Laisné
- CNRS UMR3244, Institut Curie, PSL University, Paris, France
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontorio, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontorio, Canada.
- Ontario Institute for Cancer Research, Toronto, Ontorio, Canada.
| | - Céline Vallot
- CNRS UMR3244, Institut Curie, PSL University, Paris, France.
- Translational Research Department, Institut Curie, PSL University, Paris, France.
- Single Cell Initiative, Institut Curie, PSL University, Paris, France.
| |
Collapse
|
4
|
Munir I, Nazir F, Yesiloz G. Unlocking Nature's Potential: Ferritin as a Universal Nanocarrier for Amplified Cancer Therapy Testing via 3D Microtissues. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70187-70204. [PMID: 39660468 DOI: 10.1021/acsami.4c12524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
In the existing development of extensive drug screening models, 3D cell cultures outshine conventional 2D monolayer cells by closely imitating the in vivo tumor microenvironment. This makes 3D culture a more physiologically relevant and convenient system in the regime of preclinical drug testing. In the nanomedicinal world, nanoconjugates as nanocarriers are largely hunted due to their capability of precisely binding to target cells and distributing essential dosages of therapeutic drugs with enhanced safety profiles. Thus, for boosted drug availability, the evolution from conventional drug treatment to combination therapies and last switching to drug carriers has gained significant progression in cancer cure. In contrast to conventional engineered nanoparticles, herein, we successfully designed biomolecule (ferritin)-based drug nanoconjugates effective both as a single drug (valproic acid-VPA) and twin-drug (valproic acid/doxorubicin-Dox) carriers, which dramatically enhance the proficiency of the tumor therapeutic modality. To question the reported adjuvant drug property of VPA, we progressed utilizing at first VPA alone as an effective yet exclusive tumor therapy when delivered via some carrier molecule, in particular protein. Subsequently, we paralleled this comprehensive investigation output to compare and test the coloading strategy of drugs and observe the synergistic and/or additive behavior of VPA in conjugation with other anticancer agents (Dox) while given via a carrier molecule. To approach this, VPA and/or Dox molecules were encapsulated into the ferritin (F) cavity using a thermosensitive synthesis method by maintaining the temperature at 60 °C. The successful encapsulation of drugs in the protein nanocage was confirmed through various characterization techniques. The F-VPA/F-VPA-Dox nanoconjugates exhibited similar morphology and structural characteristics to the hollow ferritin cage and showed significant cytotoxicity than the naked drugs when tested on physiologically relevant 3D spheroid models. Precisely, our first designed carrier nanoconjugate, i.e., F-VPA, offered more than a 3-fold increased intratumoral drug concentration than free VPA and significantly suppressed tumor growth after a single-dose treatment. However, our second modeled carrier nanoconjugate, viz. F-VPA-Dox, revealed an extended median survival period and lesser toxicity when administered at a much more effective dose (∼3-5 μM), in 3D tumor spheroid models of various cancer cell lines. All in all, importantly, ferritin nanoconjugates exhibited an enhanced tumor inhibition rate with a single-dose treatment, which further confirms the benefits of the active targeting property of these nanocarriers. Moreover, these nanocarriers also offer to deliver a significant dose of the therapeutic drug into tumor cells, alongside tremendous biocompatibility and safety profiles in numerous tumor 3D spheroid models.
Collapse
Affiliation(s)
- Iqra Munir
- National Nanotechnology Research Center (UNAM) Bilkent University, Cankaya, Ankara, 06800, Türkiye
| | - Faiqa Nazir
- National Nanotechnology Research Center (UNAM) Bilkent University, Cankaya, Ankara, 06800, Türkiye
- Institute of Material Science and Nanotechnology, Bilkent University, Cankaya, Ankara, 06800, Türkiye
| | - Gurkan Yesiloz
- National Nanotechnology Research Center (UNAM) Bilkent University, Cankaya, Ankara, 06800, Türkiye
- Institute of Material Science and Nanotechnology, Bilkent University, Cankaya, Ankara, 06800, Türkiye
| |
Collapse
|
5
|
Ng AM, MacKinnon KM, Cook AA, D'Alonzo RA, Rowshanfarzad P, Nowak AK, Gill S, Ebert MA. Mechanistic in silico explorations of the immunogenic and synergistic effects of radiotherapy and immunotherapy: a critical review. Phys Eng Sci Med 2024; 47:1291-1306. [PMID: 39017990 PMCID: PMC11666662 DOI: 10.1007/s13246-024-01458-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
Immunotherapy is a rapidly evolving field, with many models attempting to describe its impact on the immune system, especially when paired with radiotherapy. Tumor response to this combination involves a complex spatiotemporal dynamic which makes either clinical or pre-clinical in vivo investigation across the resulting extensive solution space extremely difficult. In this review, several in silico models of the interaction between radiotherapy, immunotherapy, and the patient's immune system are examined. The study included only mathematical models published in English that investigated the effects of radiotherapy on the immune system, or the effect of immuno-radiotherapy with immune checkpoint inhibitors. The findings indicate that treatment efficacy was predicted to improve when both radiotherapy and immunotherapy were administered, compared to radiotherapy or immunotherapy alone. However, the models do not agree on the optimal schedule and fractionation of radiotherapy and immunotherapy. This corresponds to relevant clinical trials, which report an improved treatment efficacy with combination therapy, however, the optimal scheduling varies between clinical trials. This discrepancy between the models can be attributed to the variation in model approach and the specific cancer types modeled, making the determination of the optimum general treatment schedule and model challenging. Further research needs to be conducted with similar data sets to evaluate the best model and treatment schedule for a specific cancer type and stage.
Collapse
Affiliation(s)
- Allison M Ng
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
| | - Kelly M MacKinnon
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
| | - Alistair A Cook
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Rebecca A D'Alonzo
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- Centre for Advanced Technologies in Cancer Research (CATCR), Perth, WA, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, The University of Western Australia, Crawley, WA, Australia
- Institute for Respiratory Health, Institute for Respiratory Health, Perth, WA, Australia
- Medical School, The University of Western Australia, Crawley, WA, Australia
| | - Suki Gill
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Martin A Ebert
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, WA, Australia.
- Centre for Advanced Technologies in Cancer Research (CATCR), Perth, WA, Australia.
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.
| |
Collapse
|
6
|
Updegrove TB, Delerue T, Anantharaman V, Cho H, Chan C, Nipper T, Choo-Wosoba H, Jenkins LM, Zhang L, Su Y, Shroff H, Chen J, Bewley CA, Aravind L, Ramamurthi KS. Altruistic feeding and cell-cell signaling during bacterial differentiation actively enhance phenotypic heterogeneity. SCIENCE ADVANCES 2024; 10:eadq0791. [PMID: 39423260 PMCID: PMC11488536 DOI: 10.1126/sciadv.adq0791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/12/2024] [Indexed: 10/21/2024]
Abstract
Starvation triggers bacterial spore formation, a committed differentiation program that transforms a vegetative cell into a dormant spore. Cells in a population enter sporulation nonuniformly to secure against the possibility that favorable growth conditions, which put sporulation-committed cells at a disadvantage, may resume. This heterogeneous behavior is initiated by a passive mechanism: stochastic activation of a master transcriptional regulator. Here, we identify a cell-cell communication pathway containing the proteins ShfA (YabQ) and ShfP (YvnB) that actively promotes phenotypic heterogeneity, wherein Bacillus subtilis cells that start sporulating early use a calcineurin-like phosphoesterase to release glycerol, which simultaneously acts as a signaling molecule and a nutrient to delay nonsporulating cells from entering sporulation. This produced a more diverse population that was better poised to exploit a sudden influx of nutrients compared to those generating heterogeneity via stochastic gene expression alone. Although conflict systems are prevalent among microbes, genetically encoded cooperative behavior in unicellular organisms can evidently also boost inclusive fitness.
Collapse
Affiliation(s)
- Taylor B. Updegrove
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Delerue
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivek Anantharaman
- Computational Biology Branch, Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Hyomoon Cho
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Carissa Chan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Nipper
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hyoyoung Choo-Wosoba
- Office of Collaborative Biostatistics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa M. Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lixia Zhang
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Janelia Farm Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Hari Shroff
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Janelia Farm Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Carole A. Bewley
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - L. Aravind
- Computational Biology Branch, Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Kumaran S. Ramamurthi
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Dipali SS, Gowett MQ, Kamat P, Converse A, Zaniker EJ, Fennell A, Chou T, Pritchard MT, Zelinski M, Phillip JM, Duncan FE. Self-organizing ovarian somatic organoids preserve cellular heterogeneity and reveal cellular contributions to ovarian aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607456. [PMID: 39211064 PMCID: PMC11360955 DOI: 10.1101/2024.08.10.607456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ovarian somatic cells are essential for reproductive function, but no existing ex vivo models recapitulate the cellular heterogeneity or interactions within this compartment. We engineered a novel ovarian somatic organoid model by culturing a stroma-enriched fraction of mouse ovaries in scaffold-free agarose micromolds. Ovarian somatic organoids self-organized, maintained diverse cell populations, produced extracellular matrix, and secreted hormones. Organoids generated from reproductively old mice exhibited reduced aggregation and growth compared to young counterparts, as well as differences in cellular composition. Interestingly, matrix fibroblasts from old mice demonstrated upregulation of pathways associated with the actin cytoskeleton and downregulation of cell adhesion pathways, indicative of increased cellular stiffness which may impair organoid aggregation. Cellular morphology, which is regulated by the cytoskeleton, significantly changed with age and in response to actin depolymerization. Moreover, actin depolymerization rescued age-associated organoid aggregation deficiency. Overall, ovarian somatic organoids have advanced fundamental knowledge of cellular contributions to ovarian aging.
Collapse
|
8
|
Thite NG, Tuberty-Vaughan E, Wilcox P, Wallace N, Calderon CP, Randolph TW. Stain-Free Approach to Determine and Monitor Cell Heath Using Supervised and Unsupervised Image-Based Deep Learning. J Pharm Sci 2024; 113:2114-2127. [PMID: 38710387 PMCID: PMC11670887 DOI: 10.1016/j.xphs.2024.05.001] [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: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Cell-based medicinal products (CBMPs) are a growing class of therapeutics that promise new treatments for complex and rare diseases. Given the inherent complexity of the whole human cells comprising CBMPs, there is a need for robust and fast analytical methods for characterization, process monitoring, and quality control (QC) testing during their manufacture. Existing techniques to evaluate and monitor cell quality typically constitute labor-intensive, expensive, and highly specific staining assays. In this work, we combine image-based deep learning with flow imaging microscopy (FIM) to predict cell health metrics using cellular morphology "fingerprints" extracted from images of unstained Jurkat cells (immortalized human T-lymphocyte cells). A supervised (i.e., algorithm trained with human-generated labels for images) fingerprinting algorithm, trained on images of unstained healthy and dead cells, provides a robust stain-free, non-invasive, and non-destructive method for determining cell viability. Results from the stain-free method are in good agreement with traditional stain-based cytometric viability measurements. Additionally, when trained with images of healthy cells, dead cells and cells undergoing chemically induced apoptosis, the supervised fingerprinting algorithm is able to distinguish between the three cell states, and the results are independent of specific treatments or signaling pathways. We then show that an unsupervised variational autoencoder (VAE) algorithm trained on the same images, but without human-generated labels, is able to distinguish between samples of healthy, dead and apoptotic cells along with cellular debris based on learned morphological features and without human input. With this, we demonstrate that VAEs are a powerful exploratory technique that can be used as a process monitoring analytical tool.
Collapse
Affiliation(s)
- Nidhi G Thite
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Emma Tuberty-Vaughan
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Paige Wilcox
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Nicole Wallace
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher P Calderon
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA; Ursa Analytics, Denver, CO 80212, USA
| | - Theodore W Randolph
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
9
|
Wang X, Li T, Eljilany I, Sukrithan V, Ratan A, McCarter M, Carpten J, Colman H, Ikeguchi AP, Puzanov I, Arnold S, Churchman M, Hwu P, Rodriguez PC, Dalton WS, Weiner GJ, Tarhini AA. Multicellular immune ecotypes within solid tumors predict real-world therapeutic benefits with immune checkpoint inhibitors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.19.24310726. [PMID: 39072034 PMCID: PMC11275692 DOI: 10.1101/2024.07.19.24310726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Cancer initiation, progression, and immune evasion depend on the tumor microenvironment (TME). Thus, understanding the TME immune architecture is essential for understanding tumor metastasis and therapy response. This study aimed to create an immune cell states (CSs) atlas using bulk RNA-seq data enriched by eco-type analyses to resolve the complex immune architectures in the TME. Methods We employed EcoTyper, a machine-learning (ML) framework, to study the real-world prognostic significance of immune CSs and multicellular ecosystems, utilizing molecular data from 1,610 patients with multiple malignancies who underwent immune checkpoint inhibitor (ICI) therapy within the ORIEN Avatar cohort, a well-annotated real-world dataset. Results Our analysis revealed consistent ICI-specific prognostic TME carcinoma ecotypes (CEs) (including CE1, CE9, CE10) across our pan-cancer dataset, where CE1 being more lymphocyte-deficient and CE10 being more proinflammatory. Also, the analysis of specific immune CSs across different cancers showed consistent CD8+ and CD4+ T cell CS distribution patterns. Furthermore, survival analysis of the ORIEN ICI cohort demonstrated that ecotype CE9 is associated with the most favorable survival outcomes, while CE2 is linked to the least favorable outcomes. Notably, the melanoma-specific prognostic EcoTyper model confirmed that lower predicted risk scores are associated with improved survival and better response to immunotherapy. Finally, de novo discovery of ecotypes in the ORIEN ICI dataset identified Ecotype E3 as significantly associated with poorer survival outcomes. Conclusion Our findings offer important insights into refining the patient selection process for immunotherapy in real-world practice and guiding the creation of novel therapeutic strategies to target specific ecotypes within the TME.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Tingyi Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Islam Eljilany
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Vineeth Sukrithan
- Department of Internal Medicine, Ohio State University and Arthur G James Comprehensive Cancer Center, Columbus, OH 43210 USA
| | - Aakrosh Ratan
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Martin McCarter
- Department of Surgery, University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - John Carpten
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Howard Colman
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Susanne Arnold
- Department of Medical Oncology, University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA
| | | | - Patrick Hwu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | - George J. Weiner
- Department of Internal Medicine, Carver College of Medicine, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Ahmad A. Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Pal S, Dhar R. Living in a noisy world-origins of gene expression noise and its impact on cellular decision-making. FEBS Lett 2024; 598:1673-1691. [PMID: 38724715 DOI: 10.1002/1873-3468.14898] [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: 12/21/2023] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 07/23/2024]
Abstract
The expression level of a gene can vary between genetically identical cells under the same environmental condition-a phenomenon referred to as gene expression noise. Several studies have now elucidated a central role of transcription factors in the generation of expression noise. Transcription factors, as the key components of gene regulatory networks, drive many important cellular decisions in response to cellular and environmental signals. Therefore, a very relevant question is how expression noise impacts gene regulation and influences cellular decision-making. In this Review, we summarize the current understanding of the molecular origins of expression noise, highlighting the role of transcription factors in this process, and discuss the ways in which noise can influence cellular decision-making. As advances in single-cell technologies open new avenues for studying expression noise as well as gene regulatory circuits, a better understanding of the influence of noise on cellular decisions will have important implications for many biological processes.
Collapse
Affiliation(s)
- Sampriti Pal
- Department of Bioscience and Biotechnology, IIT Kharagpur, India
| | - Riddhiman Dhar
- Department of Bioscience and Biotechnology, IIT Kharagpur, India
| |
Collapse
|
11
|
Vellan CJ, Islam T, De Silva S, Mohd Taib NA, Prasanna G, Jayapalan JJ. Exploring novel protein-based biomarkers for advancing breast cancer diagnosis: A review. Clin Biochem 2024; 129:110776. [PMID: 38823558 DOI: 10.1016/j.clinbiochem.2024.110776] [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: 02/16/2024] [Revised: 04/26/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
This review provides a contemporary examination of the evolving landscape of breast cancer (BC) diagnosis, focusing on the pivotal role of novel protein-based biomarkers. The overview begins by elucidating the multifaceted nature of BC, exploring its prevalence, subtypes, and clinical complexities. A critical emphasis is placed on the transformative impact of proteomics, dissecting the proteome to unravel the molecular intricacies of BC. Navigating through various sources of samples crucial for biomarker investigations, the review underscores the significance of robust sample processing methods and their validation in ensuring reliable outcomes. The central theme of the review revolves around the identification and evaluation of novel protein-based biomarkers. Cutting-edge discoveries are summarised, shedding light on emerging biomarkers poised for clinical application. Nevertheless, the review candidly addresses the challenges inherent in biomarker discovery, including issues of standardisation, reproducibility, and the complex heterogeneity of BC. The future direction section envisions innovative strategies and technologies to overcome existing challenges. In conclusion, the review summarises the current state of BC biomarker research, offering insights into the intricacies of proteomic investigations. As precision medicine gains momentum, the integration of novel protein-based biomarkers emerges as a promising avenue for enhancing the accuracy and efficacy of BC diagnosis. This review serves as a compass for researchers and clinicians navigating the evolving landscape of BC biomarker discovery, guiding them toward transformative advancements in diagnostic precision and personalised patient care.
Collapse
Affiliation(s)
- Christina Jane Vellan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Tania Islam
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sumadee De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Nur Aishah Mohd Taib
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Galhena Prasanna
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Jaime Jacqueline Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
Del Puerto HL, Miranda APGS, Qutob D, Ferreira E, Silva FHS, Lima BM, Carvalho BA, Roque-Souza B, Gutseit E, Castro DC, Pozzolini ET, Duarte NO, Lopes TBG, Taborda DYO, Quirino SM, Elgerbi A, Choy JS, Underwood A. Clinical Correlation of Transcription Factor SOX3 in Cancer: Unveiling Its Role in Tumorigenesis. Genes (Basel) 2024; 15:777. [PMID: 38927713 PMCID: PMC11202618 DOI: 10.3390/genes15060777] [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: 04/15/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Members of the SOX (SRY-related HMG box) family of transcription factors are crucial for embryonic development and cell fate determination. This review investigates the role of SOX3 in cancer, as aberrations in SOX3 expression have been implicated in several cancers, including osteosarcoma, breast, esophageal, endometrial, ovarian, gastric, hepatocellular carcinomas, glioblastoma, and leukemia. These dysregulations modulate key cancer outcomes such as apoptosis, epithelial-mesenchymal transition (EMT), invasion, migration, cell cycle, and proliferation, contributing to cancer development. SOX3 exhibits varied expression patterns correlated with clinicopathological parameters in diverse tumor types. This review aims to elucidate the nuanced role of SOX3 in tumorigenesis, correlating its expression with clinical and pathological characteristics in cancer patients and cellular modelsBy providing a comprehensive exploration of SOX3 involvement in cancer, this review underscores the multifaceted role of SOX3 across distinct tumor types. The complexity uncovered in SOX3 function emphasizes the need for further research to unravel its full potential in cancer therapeutics.
Collapse
Affiliation(s)
- Helen Lima Del Puerto
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Ana Paula G. S. Miranda
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Dinah Qutob
- Department of Biological Sciences, Kent State University at Stark, North Canton, OH 44720, USA;
| | - Enio Ferreira
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Felipe H. S. Silva
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Bruna M. Lima
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Barbara A. Carvalho
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Bruna Roque-Souza
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Eduardo Gutseit
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Diego C. Castro
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Emanuele T. Pozzolini
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Nayara O. Duarte
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Thacyana B. G. Lopes
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Daiana Y. O. Taborda
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Stella M. Quirino
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil (E.F.)
| | - Ahmed Elgerbi
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - John S. Choy
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Adam Underwood
- Division of Mathematics and Sciences, Walsh University, North Canton, OH 44720, USA;
| |
Collapse
|
13
|
Ginley-Hidinger M, Abewe H, Osborne K, Richey A, Kitchen N, Mortenson KL, Wissink EM, Lis J, Zhang X, Gertz J. Cis-regulatory control of transcriptional timing and noise in response to estrogen. CELL GENOMICS 2024; 4:100542. [PMID: 38663407 PMCID: PMC11099348 DOI: 10.1016/j.xgen.2024.100542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/26/2023] [Accepted: 03/27/2024] [Indexed: 05/07/2024]
Abstract
Cis-regulatory elements control transcription levels, temporal dynamics, and cell-cell variation or transcriptional noise. However, the combination of regulatory features that control these different attributes is not fully understood. Here, we used single-cell RNA-seq during an estrogen treatment time course and machine learning to identify predictors of expression timing and noise. We found that genes with multiple active enhancers exhibit faster temporal responses. We verified this finding by showing that manipulation of enhancer activity changes the temporal response of estrogen target genes. Analysis of transcriptional noise uncovered a relationship between promoter and enhancer activity, with active promoters associated with low noise and active enhancers linked to high noise. Finally, we observed that co-expression across single cells is an emergent property associated with chromatin looping, timing, and noise. Overall, our results indicate a fundamental tradeoff between a gene's ability to quickly respond to incoming signals and maintain low variation across cells.
Collapse
Affiliation(s)
- Matthew Ginley-Hidinger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Hosiana Abewe
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Kyle Osborne
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra Richey
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Noel Kitchen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Katelyn L Mortenson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Erin M Wissink
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - John Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Xiaoyang Zhang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Jason Gertz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
14
|
Updegrove TB, Delerue T, Anantharaman V, Cho H, Chan C, Nipper T, Choo-Wosoba H, Jenkins LM, Zhang L, Su Y, Shroff H, Chen J, Bewley CA, Aravind L, Ramamurthi KS. Altruistic feeding and cell-cell signaling during bacterial differentiation actively enhance phenotypic heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587046. [PMID: 38903092 PMCID: PMC11188070 DOI: 10.1101/2024.03.27.587046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Starvation triggers bacterial spore formation, a committed differentiation program that transforms a vegetative cell into a dormant spore. Cells in a population enter sporulation non-uniformly to secure against the possibility that favorable growth conditions, which puts sporulation-committed cells at a disadvantage, may resume. This heterogeneous behavior is initiated by a passive mechanism: stochastic activation of a master transcriptional regulator. Here, we identify a cell-cell communication pathway that actively promotes phenotypic heterogeneity, wherein Bacillus subtilis cells that start sporulating early utilize a calcineurin-like phosphoesterase to release glycerol, which simultaneously acts as a signaling molecule and a nutrient to delay non-sporulating cells from entering sporulation. This produced a more diverse population that was better poised to exploit a sudden influx of nutrients compared to those generating heterogeneity via stochastic gene expression alone. Although conflict systems are prevalent among microbes, genetically encoded cooperative behavior in unicellular organisms can evidently also boost inclusive fitness.
Collapse
Affiliation(s)
- Taylor B. Updegrove
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Delerue
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivek Anantharaman
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Hyomoon Cho
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Carissa Chan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Nipper
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Support Section, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa M. Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lixia Zhang
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Janelia Farm Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Hari Shroff
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Janelia Farm Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Carole A. Bewley
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - L. Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Kumaran S. Ramamurthi
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Conner SJ, Guarin JR, Le TT, Fatherree JP, Kelley C, Payne SL, Parker SR, Bloomer H, Zhang C, Salhany K, McGinn RA, Henrich E, Yui A, Srinivasan D, Borges H, Oudin MJ. Cell morphology best predicts tumorigenicity and metastasis in vivo across multiple TNBC cell lines of different metastatic potential. Breast Cancer Res 2024; 26:43. [PMID: 38468326 PMCID: PMC10929179 DOI: 10.1186/s13058-024-01796-8] [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: 06/14/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Metastasis is the leading cause of death in breast cancer patients. For metastasis to occur, tumor cells must invade locally, intravasate, and colonize distant tissues and organs, all steps that require tumor cell migration. The majority of studies on invasion and metastasis rely on human breast cancer cell lines. While it is known that these cells have different properties and abilities for growth and metastasis, the in vitro morphological, proliferative, migratory, and invasive behavior of these cell lines and their correlation to in vivo behavior is poorly understood. Thus, we sought to classify each cell line as poorly or highly metastatic by characterizing tumor growth and metastasis in a murine model of six commonly used human triple-negative breast cancer xenografts, as well as determine which in vitro assays commonly used to study cell motility best predict in vivo metastasis. METHODS We evaluated the liver and lung metastasis of human TNBC cell lines MDA-MB-231, MDA-MB-468, BT549, Hs578T, BT20, and SUM159 in immunocompromised mice. We characterized each cell line's cell morphology, proliferation, and motility in 2D and 3D to determine the variation in these parameters between cell lines. RESULTS We identified MDA-MB-231, MDA-MB-468, and BT549 cells as highly tumorigenic and metastatic, Hs578T as poorly tumorigenic and metastatic, BT20 as intermediate tumorigenic with poor metastasis to the lungs but highly metastatic to the livers, and SUM159 as intermediate tumorigenic but poorly metastatic to the lungs and livers. We showed that metrics that characterize cell morphology are the most predictive of tumor growth and metastatic potential to the lungs and liver. Further, we found that no single in vitro motility assay in 2D or 3D significantly correlated with metastasis in vivo. CONCLUSIONS Our results provide an important resource for the TNBC research community, identifying the metastatic potential of 6 commonly used cell lines. Our findings also support the use of cell morphological analysis to investigate the metastatic potential and emphasize the need for multiple in vitro motility metrics using multiple cell lines to represent the heterogeneity of metastasis in vivo.
Collapse
Affiliation(s)
- Sydney J Conner
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Justinne R Guarin
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Thanh T Le
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Jackson P Fatherree
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Charlotte Kelley
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Samantha L Payne
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON, Canada
| | - Savannah R Parker
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Crystal Zhang
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Kenneth Salhany
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Rachel A McGinn
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Emily Henrich
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Anna Yui
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Deepti Srinivasan
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Hannah Borges
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, 200 College Ave, Medford, MA, 02155, USA.
| |
Collapse
|
16
|
Bose A, Datta S, Mandal R, Ray U, Dhar R. Increased heterogeneity in expression of genes associated with cancer progression and drug resistance. Transl Oncol 2024; 41:101879. [PMID: 38262110 PMCID: PMC10832509 DOI: 10.1016/j.tranon.2024.101879] [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: 10/27/2023] [Revised: 12/16/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024] Open
Abstract
Fluctuations in the number of regulatory molecules and differences in timings of molecular events can generate variation in gene expression among genetically identical cells in the same environmental condition. This variation, termed as expression noise, can create differences in metabolic state and cellular functions, leading to phenotypic heterogeneity. Expression noise and phenotypic heterogeneity have been recognized as important contributors to intra-tumor heterogeneity, and have been associated with cancer growth, progression, and therapy resistance. However, how expression noise changes with cancer progression in actual cancer patients has remained poorly explored. Such an analysis, through identification of genes with increasing expression noise, can provide valuable insights into generation of intra-tumor heterogeneity, and could have important implications for understanding immune-suppression, drug tolerance and therapy resistance. In this work, we performed a genome-wide identification of changes in gene expression noise with cancer progression using single-cell RNA-seq data of lung adenocarcinoma patients at different stages of cancer. We identified 37 genes in epithelial cells that showed an increasing noise trend with cancer progression, many of which were also associated with cancer growth, EMT and therapy resistance. We found that expression of several of these genes was positively associated with expression of mitochondrial genes, suggesting an important role of mitochondria in generation of heterogeneity. In addition, we uncovered substantial differences in sample-specific noise profiles which could have implications for personalized prognosis and treatment.
Collapse
Affiliation(s)
- Anwesha Bose
- Department of Bioscience and Biotechnology, Indian Institute of Technology (IIT) Kharagpur, India
| | - Subhasis Datta
- Department of Bioscience and Biotechnology, Indian Institute of Technology (IIT) Kharagpur, India
| | - Rakesh Mandal
- Department of Bioscience and Biotechnology, Indian Institute of Technology (IIT) Kharagpur, India
| | - Upasana Ray
- Department of Bioscience and Biotechnology, Indian Institute of Technology (IIT) Kharagpur, India
| | - Riddhiman Dhar
- Department of Bioscience and Biotechnology, Indian Institute of Technology (IIT) Kharagpur, India.
| |
Collapse
|
17
|
Ginley-Hidinger M, Abewe H, Osborne K, Richey A, Kitchen N, Mortenson KL, Wissink EM, Lis J, Zhang X, Gertz J. Cis-regulatory control of transcriptional timing and noise in response to estrogen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.14.532457. [PMID: 36993565 PMCID: PMC10054948 DOI: 10.1101/2023.03.14.532457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cis-regulatory elements control transcription levels, temporal dynamics, and cell-cell variation or transcriptional noise. However, the combination of regulatory features that control these different attributes is not fully understood. Here, we used single cell RNA-seq during an estrogen treatment time course and machine learning to identify predictors of expression timing and noise. We find that genes with multiple active enhancers exhibit faster temporal responses. We verified this finding by showing that manipulation of enhancer activity changes the temporal response of estrogen target genes. Analysis of transcriptional noise uncovered a relationship between promoter and enhancer activity, with active promoters associated with low noise and active enhancers linked to high noise. Finally, we observed that co-expression across single cells is an emergent property associated with chromatin looping, timing, and noise. Overall, our results indicate a fundamental tradeoff between a gene's ability to quickly respond to incoming signals and maintain low variation across cells.
Collapse
Affiliation(s)
- Matthew Ginley-Hidinger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Hosiana Abewe
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Kyle Osborne
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra Richey
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Noel Kitchen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Katelyn L. Mortenson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Erin M. Wissink
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - John Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Xiaoyang Zhang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Jason Gertz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
18
|
Chanou A, Weiβ M, Holler K, Sajid A, Straub T, Krietsch J, Sanchi A, Ummethum H, Lee CSK, Kruse E, Trauner M, Werner M, Lalonde M, Lopes M, Scialdone A, Hamperl S. Single molecule MATAC-seq reveals key determinants of DNA replication origin efficiency. Nucleic Acids Res 2023; 51:12303-12324. [PMID: 37956271 PMCID: PMC10711542 DOI: 10.1093/nar/gkad1022] [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] [Received: 11/25/2022] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Stochastic origin activation gives rise to significant cell-to-cell variability in the pattern of genome replication. The molecular basis for heterogeneity in efficiency and timing of individual origins is a long-standing question. Here, we developed Methylation Accessibility of TArgeted Chromatin domain Sequencing (MATAC-Seq) to determine single-molecule chromatin accessibility of four specific genomic loci. MATAC-Seq relies on preferential modification of accessible DNA by methyltransferases combined with Nanopore-Sequencing for direct readout of methylated DNA-bases. Applying MATAC-Seq to selected early-efficient and late-inefficient yeast replication origins revealed large heterogeneity of chromatin states. Disruption of INO80 or ISW2 chromatin remodeling complexes leads to changes at individual nucleosomal positions that correlate with changes in their replication efficiency. We found a chromatin state with an accessible nucleosome-free region in combination with well-positioned +1 and +2 nucleosomes as a strong predictor for efficient origin activation. Thus, MATAC-Seq identifies the large spectrum of alternative chromatin states that co-exist on a given locus previously masked in population-based experiments and provides a mechanistic basis for origin activation heterogeneity during eukaryotic DNA replication. Consequently, our single-molecule chromatin accessibility assay will be ideal to define single-molecule heterogeneity across many fundamental biological processes such as transcription, replication, or DNA repair in vitro and ex vivo.
Collapse
Affiliation(s)
- Anna Chanou
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Matthias Weiβ
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Karoline Holler
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Atiqa Sajid
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Jana Krietsch
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Andrea Sanchi
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Henning Ummethum
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Clare S K Lee
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Elisabeth Kruse
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Manuel Trauner
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Marcel Werner
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Maxime Lalonde
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephan Hamperl
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
19
|
Wang X, Eichhorn PJA, Thiery JP. TGF-β, EMT, and resistance to anti-cancer treatment. Semin Cancer Biol 2023; 97:1-11. [PMID: 37944215 DOI: 10.1016/j.semcancer.2023.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 05/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-β-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-β on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.
Collapse
Affiliation(s)
- Xuecong Wang
- Guangzhou National Laboratory, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Pieter Johan Adam Eichhorn
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Curtin Medical School, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore, Singapore
| | | |
Collapse
|
20
|
Ozen M, Lopez CF. Data-driven structural analysis of small cell lung cancer transcription factor network suggests potential subtype regulators and transition pathways. NPJ Syst Biol Appl 2023; 9:55. [PMID: 37907529 PMCID: PMC10618210 DOI: 10.1038/s41540-023-00316-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive disease and challenging to treat due to its mixture of transcriptional subtypes and subtype transitions. Transcription factor (TF) networks have been the focus of studies to identify SCLC subtype regulators via systems approaches. Yet, their structures, which can provide clues on subtype drivers and transitions, are barely investigated. Here, we analyze the structure of an SCLC TF network by using graph theory concepts and identify its structurally important components responsible for complex signal processing, called hubs. We show that the hubs of the network are regulators of different SCLC subtypes by analyzing first the unbiased network structure and then integrating RNA-seq data as weights assigned to each interaction. Data-driven analysis emphasizes MYC as a hub, consistent with recent reports. Furthermore, we hypothesize that the pathways connecting functionally distinct hubs may control subtype transitions and test this hypothesis via network simulations on a candidate pathway and observe subtype transition. Overall, structural analyses of complex networks can identify their functionally important components and pathways driving the network dynamics. Such analyses can be an initial step for generating hypotheses and can guide the discovery of target pathways whose perturbation may change the network dynamics phenotypically.
Collapse
Affiliation(s)
- Mustafa Ozen
- Dept. of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Multiscale Modeling Group, SI3, Altos Labs, Redwood City, CA, USA
| | - Carlos F Lopez
- Dept. of Biochemistry, Vanderbilt University, Nashville, TN, USA.
- Multiscale Modeling Group, SI3, Altos Labs, Redwood City, CA, USA.
| |
Collapse
|
21
|
Salek M, Li N, Chou HP, Saini K, Jovic A, Jacobs KB, Johnson C, Lu V, Lee EJ, Chang C, Nguyen P, Mei J, Pant KP, Wong-Thai AY, Smith QF, Huang S, Chow R, Cruz J, Walker J, Chan B, Musci TJ, Ashley EA, Masaeli MM. COSMOS: a platform for real-time morphology-based, label-free cell sorting using deep learning. Commun Biol 2023; 6:971. [PMID: 37740030 PMCID: PMC10516940 DOI: 10.1038/s42003-023-05325-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
Cells are the singular building blocks of life, and a comprehensive understanding of morphology, among other properties, is crucial to the assessment of underlying heterogeneity. We developed Computational Sorting and Mapping of Single Cells (COSMOS), a platform based on Artificial Intelligence (AI) and microfluidics to characterize and sort single cells based on real-time deep learning interpretation of high-resolution brightfield images. Supervised deep learning models were applied to characterize and sort cell lines and dissociated primary tissue based on high-dimensional embedding vectors of morphology without the need for biomarker labels and stains/dyes. We demonstrate COSMOS capabilities with multiple human cell lines and tissue samples. These early results suggest that our neural networks embedding space can capture and recapitulate deep visual characteristics and can be used to efficiently purify unlabeled viable cells with desired morphological traits. Our approach resolves a technical gap in the ability to perform real-time deep learning assessment and sorting of cells based on high-resolution brightfield images.
Collapse
Affiliation(s)
- Mahyar Salek
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA.
| | - Nianzhen Li
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Hou-Pu Chou
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Kiran Saini
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Andreja Jovic
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Kevin B Jacobs
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | | | - Vivian Lu
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Esther J Lee
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | | | - Phuc Nguyen
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Jeanette Mei
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Krishna P Pant
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | | | | | | | - Ryan Chow
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Janifer Cruz
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Jeff Walker
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Bryan Chan
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Thomas J Musci
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
| | - Euan A Ashley
- Deepcell Inc; 4025 Bohannon Dr., Menlo Park, CA, 94025, USA
- Department of Medicine, Genetics, & Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
22
|
Mavropoulos A, Johnson C, Lu V, Nieto J, Schneider EC, Saini K, Phelan ML, Hsie LX, Wang MJ, Cruz J, Mei J, Kim JJ, Lian Z, Li N, Boutet SC, Wong-Thai AY, Yu W, Lu QY, Kim T, Geng Y, Masaeli MM, Lee TD, Rao J. Artificial Intelligence-Driven Morphology-Based Enrichment of Malignant Cells from Body Fluid. Mod Pathol 2023; 36:100195. [PMID: 37100228 DOI: 10.1016/j.modpat.2023.100195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/29/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023]
Abstract
Cell morphology is a fundamental feature used to evaluate patient specimens in pathologic analysis. However, traditional cytopathology analysis of patient effusion samples is limited by low tumor cell abundance coupled with the high background of nonmalignant cells, restricting the ability of downstream molecular and functional analyses to identify actionable therapeutic targets. We applied the Deepcell platform that combines microfluidic sorting, brightfield imaging, and real-time deep learning interpretations based on multidimensional morphology to enrich carcinoma cells from malignant effusions without cell staining or labels. Carcinoma cell enrichment was validated with whole genome sequencing and targeted mutation analysis, which showed a higher sensitivity for detection of tumor fractions and critical somatic variant mutations that were initially at low levels or undetectable in presort patient samples. Our study demonstrates the feasibility and added value of supplementing traditional morphology-based cytology with deep learning, multidimensional morphology analysis, and microfluidic sorting.
Collapse
Affiliation(s)
| | | | - Vivian Lu
- Deepcell, Inc, Menlo Park, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Weibo Yu
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Qing-Yi Lu
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Teresa Kim
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Yipeng Geng
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | | | - Thomas D Lee
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Jianyu Rao
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, California.
| |
Collapse
|
23
|
Thakur S, Haider S, Natrajan R. Implications of tumour heterogeneity on cancer evolution and therapy resistance: lessons from breast cancer. J Pathol 2023; 260:621-636. [PMID: 37587096 DOI: 10.1002/path.6158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 08/18/2023]
Abstract
Tumour heterogeneity is pervasive amongst many cancers and leads to disease progression, and therapy resistance. In this review, using breast cancer as an exemplar, we focus on the recent advances in understanding the interplay between tumour cells and their microenvironment using single cell sequencing and digital spatial profiling technologies. Further, we discuss the utility of lineage tracing methodologies in pre-clinical models of breast cancer, and how these are being used to unravel new therapeutic vulnerabilities and reveal biomarkers of breast cancer progression. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Shefali Thakur
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| |
Collapse
|
24
|
Sunnerberg J, Thomas WS, Petusseau A, Reed MS, Jack Hoopes P, Pogue BW. Review of optical reporters of radiation effects in vivo: tools to quantify improvements in radiation delivery technique. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:080901. [PMID: 37560327 PMCID: PMC10409499 DOI: 10.1117/1.jbo.28.8.080901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
Significance Radiation damage studies are used to optimize radiotherapy treatment techniques. Although biological indicators of damage are the best assays of effect, they are highly variable due to biological heterogeneity. The free radical radiochemistry can be assayed with optical reporters, allowing for high precision titration of techniques. Aim We examine the optical reporters of radiochemistry to highlight those with the best potential for translational use in vivo, as surrogates for biological damage assays, to inform on mechanisms. Approach A survey of the radical chemistry effects from reactive oxygen species (ROS) and oxygen itself was completed to link to DNA or biological damage. Optical reporters of ROS include fluorescent, phosphorescent, and bioluminescent molecules that have a variety of activation pathways, and each was reviewed for its in vivo translation potential. Results There are molecular reporters of ROS having potential to report within living systems, including derivatives of luminol, 2'7'-dichlorofluorescein diacetate, Amplex Red, and fluorescein. None have unique specificity to singular ROS species. Macromolecular engineered reporters unique to specific ROS are emerging. The ability to directly measure oxygen via reporters, such as Oxyphor and protoporphyrin IX, is an opportunity to quantify the consumption of oxygen during ROS generation, and this translates from in vitro to in vivo use. Emerging techniques, such as ion particle beams, spatial fractionation, and ultra-high dose rate FLASH radiotherapy, provide the motivation for these studies. Conclusions In vivo optical reporters of radiochemistry are quantitatively useful for comparing radiotherapy techniques, although their use comes at the cost of the unknown connection to the mechanisms of radiobiological damage. Still their lower measurement uncertainty, compared with biological response assay, makes them an invaluable tool. Linkage to DNA damage and biological damage is needed, and measures such as oxygen consumption serve as useful surrogate measures that translate to in vivo use.
Collapse
Affiliation(s)
- Jacob Sunnerberg
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - William S. Thomas
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Arthur Petusseau
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Matthew S. Reed
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - P. Jack Hoopes
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Brian W. Pogue
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| |
Collapse
|
25
|
Wan Y, Cohen J, Szenk M, Farquhar KS, Coraci D, Krzysztoń R, Azukas J, Van Nest N, Smashnov A, Chern YJ, De Martino D, Nguyen LC, Bien H, Bravo-Cordero JJ, Chan CH, Rosner MR, Balázsi G. Nonmonotone invasion landscape by noise-aware control of metastasis activator levels. Nat Chem Biol 2023; 19:887-899. [PMID: 37231268 PMCID: PMC10299915 DOI: 10.1038/s41589-023-01344-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/18/2023] [Indexed: 05/27/2023]
Abstract
A major pharmacological assumption is that lowering disease-promoting protein levels is generally beneficial. For example, inhibiting metastasis activator BACH1 is proposed to decrease cancer metastases. Testing such assumptions requires approaches to measure disease phenotypes while precisely adjusting disease-promoting protein levels. Here we developed a two-step strategy to integrate protein-level tuning, noise-aware synthetic gene circuits into a well-defined human genomic safe harbor locus. Unexpectedly, engineered MDA-MB-231 metastatic human breast cancer cells become more, then less and then more invasive as we tune BACH1 levels up, irrespective of the native BACH1. BACH1 expression shifts in invading cells, and expression of BACH1's transcriptional targets confirm BACH1's nonmonotone phenotypic and regulatory effects. Thus, chemical inhibition of BACH1 could have unwanted effects on invasion. Additionally, BACH1's expression variability aids invasion at high BACH1 expression. Overall, precisely engineered, noise-aware protein-level control is necessary and important to unravel disease effects of genes to improve clinical drug efficacy.
Collapse
Affiliation(s)
- Yiming Wan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Joseph Cohen
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Mariola Szenk
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Kevin S Farquhar
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center, UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Damiano Coraci
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Rafał Krzysztoń
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Joshua Azukas
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Nicholas Van Nest
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Alex Smashnov
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Yi-Jye Chern
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Daniela De Martino
- Department of Medicine, Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Chi Nguyen
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Harold Bien
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chia-Hsin Chan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Gábor Balázsi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA.
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
26
|
Nowak CM, Quarton T, Bleris L. Impact of variability in cell cycle periodicity on cell population dynamics. PLoS Comput Biol 2023; 19:e1011080. [PMID: 37339124 DOI: 10.1371/journal.pcbi.1011080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/06/2023] [Indexed: 06/22/2023] Open
Abstract
The cell cycle consists of a series of orchestrated events controlled by molecular sensing and feedback networks that ultimately drive the duplication of total DNA and the subsequent division of a single parent cell into two daughter cells. The ability to block the cell cycle and synchronize cells within the same phase has helped understand factors that control cell cycle progression and the properties of each individual phase. Intriguingly, when cells are released from a synchronized state, they do not maintain synchronized cell division and rapidly become asynchronous. The rate and factors that control cellular desynchronization remain largely unknown. In this study, using a combination of experiments and simulations, we investigate the desynchronization properties in cervical cancer cells (HeLa) starting from the G1/S boundary following double-thymidine block. Propidium iodide (PI) DNA staining was used to perform flow cytometry cell cycle analysis at regular 8 hour intervals, and a custom auto-similarity function to assess the desynchronization and quantify the convergence to an asynchronous state. In parallel, we developed a single-cell phenomenological model the returns the DNA amount across the cell cycle stages and fitted the parameters using experimental data. Simulations of population of cells reveal that the cell cycle desynchronization rate is primarily sensitive to the variability of cell cycle duration within a population. To validate the model prediction, we introduced lipopolysaccharide (LPS) to increase cell cycle noise. Indeed, we observed an increase in cell cycle variability under LPS stimulation in HeLa cells, accompanied with an enhanced rate of cell cycle desynchronization. Our results show that the desynchronization rate of artificially synchronized in-phase cell populations can be used a proxy of the degree of variance in cell cycle periodicity, an underexplored axis in cell cycle research.
Collapse
Affiliation(s)
- Chance M Nowak
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, United States of America
| | - Tyler Quarton
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
| | - Leonidas Bleris
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, United States of America
| |
Collapse
|
27
|
Conner S, Guarin JR, Le TT, Fatherree J, Kelley C, Payne S, Salhany K, McGinn R, Henrich E, Yui A, Parker S, Srinivasan D, Bloomer H, Borges H, Oudin MJ. Cell morphology best predicts tumorigenicity and metastasis in vivo across multiple TNBC cell lines of different metastatic potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.544969. [PMID: 37398306 PMCID: PMC10312673 DOI: 10.1101/2023.06.14.544969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Metastasis is the leading cause of death in breast cancer patients. For metastasis to occur, tumor cells must invade locally, intravasate, and colonize distant tissues and organs, all steps that require tumor cell migration. The majority of studies on invasion and metastasis rely on human breast cancer cell lines. While it is known that these cells have different properties and abilities for growth and metastasis, the in vitro morphological, proliferative, migratory, and invasive behavior of these cell lines and their correlation to in vivo behavior is poorly understood. Thus, we sought to classify each cell line as poorly or highly metastatic by characterizing tumor growth and metastasis in a murine model of six commonly used human triple-negative breast cancer xenografts, as well as determine which in vitro assays commonly used to study cell motility best predict in vivo metastasis. Methods We evaluated the liver and lung metastasis of human TNBC cell lines MDA-MB-231, MDA-MB-468, BT549, Hs578T, BT20, and SUM159 in immunocompromised mice. We characterized each cell line's cell morphology, proliferation, and motility in 2D and 3D to determine the variation in these parameters between cell lines. Results We identified MDA-MB-231, MDA-MB-468, and BT549 cells as highly tumorigenic and metastatic, Hs578T as poorly tumorigenic and metastatic, BT20 as intermediate tumorigenic with poor metastasis to the lungs but highly metastatic to the livers, and SUM159 as intermediate tumorigenic but poorly metastatic to the lungs and livers. We showed that metrics that characterize cell morphology are the most predictive of tumor growth and metastatic potential to the lungs and liver. Further, we found that no single in vitro motility assay in 2D or 3D significantly correlated with metastasis in vivo. Conclusions Our results provide an important resource for the TNBC research community, identifying the metastatic potential of 6 commonly used cell lines. Our findings also support the use of cell morphological analysis to investigate the metastatic potential and emphasize the need for multiple in vitro motility metrics using multiple cell lines to represent the heterogeneity of metastasis in vivo.
Collapse
Affiliation(s)
- Sydney Conner
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Justinne R. Guarin
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Thanh T. Le
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | | | - Charlotte Kelley
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Samantha Payne
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Ken Salhany
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Rachel McGinn
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Emily Henrich
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Anna Yui
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Savannah Parker
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Deepti Srinivasan
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Hanan Bloomer
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Hannah Borges
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, USA
| |
Collapse
|
28
|
Ozen M, Lopez CF. Data-driven structural analysis of Small Cell Lung Cancer transcription factor network suggests potential subtype regulators and transition pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.01.535226. [PMID: 37066351 PMCID: PMC10104011 DOI: 10.1101/2023.04.01.535226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Small Cell Lung Cancer (SCLC) is an aggressive disease and challenging to treat due to its mixture of transcriptional subtypes and subtype transitions. Transcription factor (TF) networks have been the focus of studies to identify SCLC subtype regulators via systems approaches. Yet, their structures, which can provide clues on subtype drivers and transitions, are barely investigated. Here, we analyze the structure of an SCLC TF network by using graph theory concepts and identify its structurally important components responsible for complex signal processing, called hubs. We show that the hubs of the network are regulators of different SCLC subtypes by analyzing first the unbiased network structure and then integrating RNA-seq data as weights assigned to each interaction. Data-driven analysis emphasizes MYC as a hub, consistent with recent reports. Furthermore, we hypothesize that the pathways connecting functionally distinct hubs may control subtype transitions and test this hypothesis via network simulations on a candidate pathway and observe subtype transition. Overall, structural analyses of complex networks can identify their functionally important components and pathways driving the network dynamics. Such analyses can be an initial step for generating hypotheses and can guide the discovery of target pathways whose perturbation may change the network dynamics phenotypically.
Collapse
Affiliation(s)
- Mustafa Ozen
- Dept. of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
- Currently at: Computational Innovation Hub, Multiscale Modeling Group, Altos Labs, Redwood City, CA 94065, USA
| | - Carlos F. Lopez
- Dept. of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
- Currently at: Computational Innovation Hub, Multiscale Modeling Group, Altos Labs, Redwood City, CA 94065, USA
| |
Collapse
|
29
|
Im Y, Kim Y. A Comprehensive Overview of RNA Deconvolution Methods and Their Application. Mol Cells 2023; 46:99-105. [PMID: 36859474 PMCID: PMC9982058 DOI: 10.14348/molcells.2023.2178] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 03/03/2023] Open
Abstract
Tumors are surrounded by a variety of tumor microenvironmental cells. Profiling individual cells within the tumor tissues is crucial to characterize the tumor microenvironment and its therapeutic implications. Since single-cell technologies are still not cost-effective, scientists have developed many statistical deconvolution methods to delineate cellular characteristics from bulk transcriptome data. Here, we present an overview of 20 deconvolution techniques, including cutting-edge techniques recently established. We categorized deconvolution techniques by three primary criteria: characteristics of methodology, use of prior knowledge of cell types and outcome of the methods. We highlighted the advantage of the recent deconvolution tools that are based on probabilistic models. Moreover, we illustrated two scenarios of the common application of deconvolution methods to study tumor microenvironments. This comprehensive review will serve as a guideline for the researchers to select the appropriate method for their application of deconvolution.
Collapse
Affiliation(s)
- Yebin Im
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Yongsoo Kim
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
30
|
Tong JH, Elmore S, Huang SS, Tachachartvanich P, Manz K, Pennell K, Wilson MD, Borowsky A, La Merrill MA. Chronic Exposure to Low Levels of Parabens Increases Mammary Cancer Growth and Metastasis in Mice. Endocrinology 2023; 164:bqad007. [PMID: 36683225 PMCID: PMC10205179 DOI: 10.1210/endocr/bqad007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/24/2023]
Abstract
Methylparaben (MP) and propylparaben (PP) are commonly used as food, cosmetic, and drug preservatives. These parabens are detected in the majority of US women and children, bind and activate estrogen receptors (ER), and stimulate mammary tumor cell growth and invasion in vitro. Hemizygous B6.FVB-Tg (MMTV-PyVT)634Mul/LellJ female mice (n = 20/treatment) were exposed to MP or PP at levels within the US Food and Drug Administration's "human acceptable daily intake." These paraben-exposed mice had increased mammary tumor volume compared with control mice (P < 0.001) and a 28% and 91% increase in the number of pulmonary metastases per week compared with the control mice, respectively (P < 0.0001). MP and PP caused differential expression of 288 and 412 mammary tumor genes, respectively (false discovery rate < 0.05), a subset of which has been associated with human breast cancer metastasis. Molecular docking and luciferase reporter studies affirmed that MP and PP bound and activated human ER, and RNA-sequencing revealed increased ER expression in mammary tumors among paraben-exposed mice. However, ER signaling was not enriched in mammary tumors. Instead, both parabens strongly impaired tumor RNA metabolism (eg, ribosome, spliceosome), as evident from enriched KEGG pathway analysis of differential mammary tumor gene expression common to both paraben treatments (MP, P < 0.001; PP, P < 0.01). Indeed, mammary tumors from PP-exposed mice had an increased retention of introns (P < 0.05). Our data suggest that parabens cause substantial mammary cancer metastasis in mice as a function of their increasing alkyl chain length and highlight the emerging role of aberrant spliceosome activity in breast cancer metastasis.
Collapse
Affiliation(s)
- Jason H Tong
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Sarah Elmore
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Shenq-Shyang Huang
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Phum Tachachartvanich
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Katherine Manz
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kurt Pennell
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Machelle D Wilson
- Department of Public Health Sciences, University of California at Davis, Davis, CA 95616, USA
| | - Alexander Borowsky
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Michele A La Merrill
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| |
Collapse
|
31
|
Chan TJ, Zhang X, Mak M. Biophysical informatics reveals distinctive phenotypic signatures and functional diversity of single-cell lineages. Bioinformatics 2023; 39:6969104. [PMID: 36610710 PMCID: PMC9825265 DOI: 10.1093/bioinformatics/btac833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022] Open
Abstract
MOTIVATION In this work, we present an analytical method for quantifying both single-cell morphologies and cell network topologies of tumor cell populations and use it to predict 3D cell behavior. RESULTS We utilized a supervised deep learning approach to perform instance segmentation on label-free live cell images across a wide range of cell densities. We measured cell shape properties and characterized network topologies for 136 single-cell clones derived from the YUMM1.7 and YUMMER1.7 mouse melanoma cell lines. Using an unsupervised clustering algorithm, we identified six distinct morphological subclasses. We further observed differences in tumor growth and invasion dynamics across subclasses in an in vitro 3D spheroid model. Compared to existing methods for quantifying 2D or 3D phenotype, our analytical method requires less time, needs no specialized equipment and is capable of much higher throughput, making it ideal for applications such as high-throughput drug screening and clinical diagnosis. AVAILABILITY AND IMPLEMENTATION https://github.com/trevor-chan/Melanoma_NetworkMorphology. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Trevor J Chan
- Department of Bioengineering, Yale University, New Haven, CT 06511, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xingjian Zhang
- Department of Bioengineering, Yale University, New Haven, CT 06511, USA
| | | |
Collapse
|
32
|
Andrade de Oliveira K, Sengupta S, Yadav AK, Clarke R. The complex nature of heterogeneity and its roles in breast cancer biology and therapeutic responsiveness. Front Endocrinol (Lausanne) 2023; 14:1083048. [PMID: 36909339 PMCID: PMC9997040 DOI: 10.3389/fendo.2023.1083048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Heterogeneity is a complex feature of cells and tissues with many interacting components. Depending on the nature of the research context, interacting features of cellular, drug response, genetic, molecular, spatial, temporal, and vascular heterogeneity may be present. We describe the various forms of heterogeneity with examples of their interactions and how they play a role in affecting cellular phenotype and drug responses in breast cancer. While cellular heterogeneity may be the most widely described and invoked, many forms of heterogeneity are evident within the tumor microenvironment and affect responses to the endocrine and cytotoxic drugs widely used in standard clinical care. Drug response heterogeneity is a critical determinant of clinical response and curative potential and also is multifaceted when encountered. The interactive nature of some forms of heterogeneity is readily apparent. For example, the process of metastasis has the properties of both temporal and spatial heterogeneity within the host, whereas each individual metastatic deposit may exhibit cellular, genetic, molecular, and vascular heterogeneity. This review describes the many forms of heterogeneity, their integrated activities, and offers some insights into how heterogeneity may be understood and studied in the future.
Collapse
Affiliation(s)
- Karla Andrade de Oliveira
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- Department of Biochemistry and Pharmacology, Universidade Federal do Piaui, Piauí, Brazil
| | - Surojeet Sengupta
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- *Correspondence: Robert Clarke,
| |
Collapse
|
33
|
O'Neill H, Lee H, Gupta I, Rodger EJ, Chatterjee A. Single-Cell DNA Methylation Analysis in Cancer. Cancers (Basel) 2022; 14:6171. [PMID: 36551655 PMCID: PMC9777108 DOI: 10.3390/cancers14246171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Morphological, transcriptomic, and genomic defects are well-explored parameters of cancer biology. In more recent years, the impact of epigenetic influences, such as DNA methylation, is becoming more appreciated. Aberrant DNA methylation has been implicated in many types of cancers, influencing cell type, state, transcriptional regulation, and genomic stability to name a few. Traditionally, large populations of cells from the tissue of interest are coalesced for analysis, producing averaged methylome data. Considering the inherent heterogeneity of cancer, analysing populations of cells as a whole denies the ability to discover novel aberrant methylation patterns, identify subpopulations, and trace cell lineages. Due to recent advancements in technology, it is now possible to obtain methylome data from single cells. This has both research and clinical implications, ranging from the identification of biomarkers to improved diagnostic tools. As with all emerging technologies, distinct experimental, bioinformatic, and practical challenges present themselves. This review begins with exploring the potential impact of single-cell sequencing on understanding cancer biology and how it could eventually benefit a clinical setting. Following this, the techniques and experimental approaches which made this technology possible are explored. Finally, the present challenges currently associated with single-cell DNA methylation sequencing are described.
Collapse
Affiliation(s)
- Hannah O'Neill
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Heather Lee
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Euan J Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
- School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Dehradun 248007, India
| |
Collapse
|
34
|
Parab L, Pal S, Dhar R. Transcription factor binding process is the primary driver of noise in gene expression. PLoS Genet 2022; 18:e1010535. [PMID: 36508455 PMCID: PMC9779669 DOI: 10.1371/journal.pgen.1010535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/22/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
Noise in expression of individual genes gives rise to variations in activity of cellular pathways and generates heterogeneity in cellular phenotypes. Phenotypic heterogeneity has important implications for antibiotic persistence, mutation penetrance, cancer growth and therapy resistance. Specific molecular features such as the presence of the TATA box sequence and the promoter nucleosome occupancy have been associated with noise. However, the relative importance of these features in noise regulation is unclear and how well these features can predict noise has not yet been assessed. Here through an integrated statistical model of gene expression noise in yeast we found that the number of regulating transcription factors (TFs) of a gene was a key predictor of noise, whereas presence of the TATA box and the promoter nucleosome occupancy had poor predictive power. With an increase in the number of regulatory TFs, there was a rise in the number of cooperatively binding TFs. In addition, an increased number of regulatory TFs meant more overlaps in TF binding sites, resulting in competition between TFs for binding to the same region of the promoter. Through modeling of TF binding to promoter and application of stochastic simulations, we demonstrated that competition and cooperation among TFs could increase noise. Thus, our work uncovers a process of noise regulation that arises out of the dynamics of gene regulation and is not dependent on any specific transcription factor or specific promoter sequence.
Collapse
Affiliation(s)
- Lavisha Parab
- Department of Biotechnology, Indian Institute of Technology (IIT) Kharagpur, Kharagpur, West Bengal, India
- Max-Planck-Institute for Evolutionary Biology, Plön, Germany
| | - Sampriti Pal
- Department of Biotechnology, Indian Institute of Technology (IIT) Kharagpur, Kharagpur, West Bengal, India
| | - Riddhiman Dhar
- Department of Biotechnology, Indian Institute of Technology (IIT) Kharagpur, Kharagpur, West Bengal, India
- * E-mail:
| |
Collapse
|
35
|
Hu X, Wu M, Ma T, Zhang Y, Zou C, Wang R, Zhang Y, Ren Y, Li Q, Liu H, Li H, Wang T, Sun X, Yang Y, Tang M, Li X, Li J, Gao X, Li T, Zhou X. Single-cell transcriptomics reveals distinct cell response between acute and chronic pulmonary infection of Pseudomonas aeruginosa. MedComm (Beijing) 2022; 3:e193. [PMID: 36514779 PMCID: PMC9732387 DOI: 10.1002/mco2.193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
Knowledge of the changes in the immune microenvironment during pulmonary bacterial acute and chronic infections is limited. The dissection of immune system may provide a basis for effective therapeutic strategies against bacterial infection. Here, we describe a single immune cell atlas of mouse lungs after acute and chronic Pseudomonas aeruginosa infection using single-cell transcriptomics, multiplex immunohistochemistry, and flow cytometry. Our single-cell transcriptomic analysis revealed large-scale comprehensive changes in immune cell composition and high variation in cell-cell interactions after acute and chronic P. aeruginosa infection. Bacterial infection reprograms the genetic architecture of immune cell populations. We identified specific immune cell types, including Cxcl2+ B cells and interstitial macrophages, in response to acute and chronic infection, such as their proportions, distribution, and functional status. Importantly, the patterns of immune cell response are drastically different between acute and chronic infection models. The distinct molecular signatures highlight the importance of the highly dynamic cell-cell interaction process in different pathological conditions, which has not been completely revealed previously. These findings provide a comprehensive and unbiased immune cellular landscape for respiratory pathogenesis in mice, enabling further understanding of immunologic mechanisms in infection and inflammatory diseases.
Collapse
Affiliation(s)
- Xueli Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Mingbo Wu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Teng Ma
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yige Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Chaoyu Zou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Ruihuan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yongxin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yuan Ren
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Qianqian Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Huan Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Heyue Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Taolin Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xiaolong Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Miao Tang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xuefeng Li
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jing Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xiang Gao
- Department of Neurosurgery and Institute of NeurosurgeryState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Taiwen Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| |
Collapse
|
36
|
Tumor-promoting aftermath post-chemotherapy: A focus on breast cancer. Life Sci 2022; 310:121125. [DOI: 10.1016/j.lfs.2022.121125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022]
|
37
|
Earnest-Noble LB, Hsu D, Chen S, Asgharian H, Nandan M, Passarelli MC, Goodarzi H, Tavazoie SF. Two isoleucyl tRNAs that decode synonymous codons divergently regulate breast cancer metastatic growth by controlling translation of proliferation-regulating genes. NATURE CANCER 2022; 3:1484-1497. [PMID: 36510010 PMCID: PMC11323107 DOI: 10.1038/s43018-022-00469-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
The human genome contains 61 codons encoding 20 amino acids. Synonymous codons representing a given amino acid are decoded by a set of transfer RNAs (tRNAs) called isoacceptors. We report the surprising observation that two isoacceptor tRNAs that decode synonymous codons become modulated in opposing directions during breast cancer progression. Specifically, tRNAIleUAU became upregulated, whereas tRNAIleGAU became repressed as breast cancer cells attained enhanced metastatic capacity. Functionally, tRNAIleUAU promoted and tRNAIleGAU suppressed metastatic colonization in mouse xenograft models. These tRNAs mediated opposing effects on codon-dependent translation of growth-promoting genes, consistent with genomic enrichment or depletion of their cognate codons in mitotic genes. Our findings uncover a specific isoacceptor tRNA pair that act in opposition, divergently impacting growth-regulating genes and a disease phenotype. Degeneracy of the genetic code can thus be biologically exploited by human cancer cells via tRNA isoacceptor shifts that causally facilitate the transition toward a growth-promoting state.
Collapse
Affiliation(s)
- Lisa B Earnest-Noble
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Dennis Hsu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Siyu Chen
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hosseinali Asgharian
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Mandayam Nandan
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Maria C Passarelli
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
38
|
Urbanski L, Brugiolo M, Park S, Angarola BL, Leclair NK, Yurieva M, Palmer P, Sahu SK, Anczuków O. MYC regulates a pan-cancer network of co-expressed oncogenic splicing factors. Cell Rep 2022; 41:111704. [DOI: 10.1016/j.celrep.2022.111704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 05/16/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
|
39
|
Nevarez AJ, Hao N. Quantitative cell imaging approaches to metastatic state profiling. Front Cell Dev Biol 2022; 10:1048630. [PMID: 36393865 PMCID: PMC9640958 DOI: 10.3389/fcell.2022.1048630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic heterogeneity of metastatic dissemination has proven challenging to identify exploitable markers of metastasis; this bottom-up approach has caused a stalemate between advances in metastasis and the late stage of the disease. Advancements in quantitative cellular imaging have allowed the detection of morphological phenotype changes specific to metastasis, the morphological changes connected to the underlying complex signaling pathways, and a robust readout of metastatic cell state. This review focuses on the recent machine and deep learning developments to gain detailed information about the metastatic cell state using light microscopy. We describe the latest studies using quantitative cell imaging approaches to identify cell appearance-based metastatic patterns. We discuss how quantitative cancer biologists can use these frameworks to work backward toward exploitable hidden drivers in the metastatic cascade and pioneering new Frontier drug discoveries specific for metastasis.
Collapse
Affiliation(s)
| | - Nan Hao
- *Correspondence: Andres J. Nevarez, ; Nan Hao,
| |
Collapse
|
40
|
García-Sancha N, Corchado-Cobos R, Gómez-Vecino A, Jiménez-Navas A, Pérez-Baena MJ, Blanco-Gómez A, Holgado-Madruga M, Mao JH, Cañueto J, Castillo-Lluva S, Mendiburu-Eliçabe M, Pérez-Losada J. Evolutionary Origins of Metabolic Reprogramming in Cancer. Int J Mol Sci 2022; 23:12063. [PMID: 36292921 PMCID: PMC9603151 DOI: 10.3390/ijms232012063] [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: 08/24/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic changes that facilitate tumor growth are one of the hallmarks of cancer. These changes are not specific to tumors but also take place during the physiological growth of tissues. Indeed, the cellular and tissue mechanisms present in the tumor have their physiological counterpart in the repair of tissue lesions and wound healing. These molecular mechanisms have been acquired during metazoan evolution, first to eliminate the infection of the tissue injury, then to enter an effective regenerative phase. Cancer itself could be considered a phenomenon of antagonistic pleiotropy of the genes involved in effective tissue repair. Cancer and tissue repair are complex traits that share many intermediate phenotypes at the molecular, cellular, and tissue levels, and all of these are integrated within a Systems Biology structure. Complex traits are influenced by a multitude of common genes, each with a weak effect. This polygenic component of complex traits is mainly unknown and so makes up part of the missing heritability. Here, we try to integrate these different perspectives from the point of view of the metabolic changes observed in cancer.
Collapse
Affiliation(s)
- Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Aurora Gómez-Vecino
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Alejandro Jiménez-Navas
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Manuel Jesús Pérez-Baena
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Marina Holgado-Madruga
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain
| | - Jian-Hua Mao
- Lawrence Berkeley National Laboratory, Biological Systems and Engineering Division, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Javier Cañueto
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Dermatología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Marina Mendiburu-Eliçabe
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
41
|
Wong KS, Zhong X, Low CSL, Kanchanawong P. Self-supervised classification of subcellular morphometric phenotypes reveals extracellular matrix-specific morphological responses. Sci Rep 2022; 12:15329. [PMID: 36097150 PMCID: PMC9468179 DOI: 10.1038/s41598-022-19472-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cell morphology is profoundly influenced by cellular interactions with microenvironmental factors such as the extracellular matrix (ECM). Upon adhesion to specific ECM, various cell types are known to exhibit different but distinctive morphologies, suggesting that ECM-dependent cell morphological responses may harbour rich information on cellular signalling states. However, the inherent morphological complexity of cellular and subcellular structures has posed an ongoing challenge for automated quantitative analysis. Since multi-channel fluorescence microscopy provides robust molecular specificity important for the biological interpretations of observed cellular architecture, here we develop a deep learning-based analysis pipeline for the classification of cell morphometric phenotypes from multi-channel fluorescence micrographs, termed SE-RNN (residual neural network with squeeze-and-excite blocks). We demonstrate SERNN-based classification of distinct morphological signatures observed when fibroblasts or epithelial cells are presented with different ECM. Our results underscore how cell shapes are non-random and established the framework for classifying cell shapes into distinct morphological signature in a cell-type and ECM-specific manner.
Collapse
Affiliation(s)
- Kin Sun Wong
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Xueying Zhong
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Christine Siok Lan Low
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Pakorn Kanchanawong
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Republic of Singapore. .,Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore.
| |
Collapse
|
42
|
Zatulovskiy E, Lanz MC, Zhang S, McCarthy F, Elias JE, Skotheim JM. Delineation of proteome changes driven by cell size and growth rate. Front Cell Dev Biol 2022; 10:980721. [PMID: 36133920 PMCID: PMC9483106 DOI: 10.3389/fcell.2022.980721] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/09/2022] [Indexed: 01/10/2023] Open
Abstract
Increasing cell size drives changes to the proteome, which affects cell physiology. As cell size increases, some proteins become more concentrated while others are diluted. As a result, the state of the cell changes continuously with increasing size. In addition to these proteomic changes, large cells have a lower growth rate (protein synthesis rate per unit volume). That both the cell's proteome and growth rate change with cell size suggests they may be interdependent. To test this, we used quantitative mass spectrometry to measure how the proteome changes in response to the mTOR inhibitor rapamycin, which decreases the cellular growth rate and has only a minimal effect on cell size. We found that large cell size and mTOR inhibition, both of which lower the growth rate of a cell, remodel the proteome in similar ways. This suggests that many of the effects of cell size are mediated by the size-dependent slowdown of the cellular growth rate. For example, the previously reported size-dependent expression of some senescence markers could reflect a cell's declining growth rate rather than its size per se. In contrast, histones and other chromatin components are diluted in large cells independently of the growth rate, likely so that they remain in proportion with the genome. Finally, size-dependent changes to the cell's growth rate and proteome composition are still apparent in cells continually exposed to a saturating dose of rapamycin, which indicates that cell size can affect the proteome independently of mTORC1 signaling. Taken together, our results clarify the dependencies between cell size, growth, mTOR activity, and the proteome remodeling that ultimately controls many aspects of cell physiology.
Collapse
Affiliation(s)
| | - Michael C. Lanz
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, Stanford, CA, United States
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, United States
| | | | | | - Jan M. Skotheim
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, Stanford, CA, United States
| |
Collapse
|
43
|
Zhang S, Zatulovskiy E, Arand J, Sage J, Skotheim JM. The cell cycle inhibitor RB is diluted in G1 and contributes to controlling cell size in the mouse liver. Front Cell Dev Biol 2022; 10:965595. [PMID: 36092730 PMCID: PMC9452963 DOI: 10.3389/fcell.2022.965595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 12/14/2022] Open
Abstract
Every type of cell in an animal maintains a specific size, which likely contributes to its ability to perform its physiological functions. While some cell size control mechanisms are beginning to be elucidated through studies of cultured cells, it is unclear if and how such mechanisms control cell size in an animal. For example, it was recently shown that RB, the retinoblastoma protein, was diluted by cell growth in G1 to promote size-dependence of the G1/S transition. However, it remains unclear to what extent the RB-dilution mechanism controls cell size in an animal. We therefore examined the contribution of RB-dilution to cell size control in the mouse liver. Consistent with the RB-dilution model, genetic perturbations decreasing RB protein concentrations through inducible shRNA expression or through liver-specific Rb1 knockout reduced hepatocyte size, while perturbations increasing RB protein concentrations in an Fah -/- mouse model increased hepatocyte size. Moreover, RB concentration reflects cell size in G1 as it is lower in larger G1 hepatocytes. In contrast, concentrations of the cell cycle activators Cyclin D1 and E2f1 were relatively constant. Lastly, loss of Rb1 weakened cell size control, i.e., reduced the inverse correlation between how much cells grew in G1 and how large they were at birth. Taken together, our results show that an RB-dilution mechanism contributes to cell size control in the mouse liver by linking cell growth to the G1/S transition.
Collapse
Affiliation(s)
- Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, United States
| | | | - Julia Arand
- Departments of Pediatrics and Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Julien Sage
- Departments of Pediatrics and Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jan M. Skotheim
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
44
|
Boguszewicz Ł. Predictive Biomarkers for Response and Toxicity of Induction Chemotherapy in Head and Neck Cancers. Front Oncol 2022; 12:900903. [PMID: 35875133 PMCID: PMC9299243 DOI: 10.3389/fonc.2022.900903] [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: 03/21/2022] [Accepted: 05/24/2022] [Indexed: 01/17/2023] Open
Abstract
This review focuses on the molecular biology of head and neck squamous cell carcinomas and presents current and emerging biomarkers of the response of patients to induction chemotherapy. The usefulness of genes, proteins, and parameters from diagnostic clinical imaging as well as other clinicopathological parameters is thoroughly discussed. The role of induction chemotherapy before radiotherapy or before chemo-radiotherapy is still debated, as the data on its efficacy are somehow confusing. Despite the constant improvement of treatment protocols and the introduction of new cytostatics, there is still no consensus regarding the use of induction chemotherapy in the treatment of head and neck cancer, with the possible exception of larynx preservation. Such difficulties indicate that potential future treatment strategies should be personalized. Personalized medicine, in which individual tumor genetics drive the selection of targeted therapies and treatment plans for each patient, has recently emerged as the next generation of cancer therapy. Early prediction of treatment outcome or its toxicity may be highly beneficial for those who are at risk of the development of severe toxicities or treatment failure—a different treatment strategy may be applied to these patients, sparing them unnecessary pain. The literature search was carried out in the PubMed and ScienceDirect databases as well as in the selected conference proceedings repositories. Of the 265 articles and abstracts found, only 30 met the following inclusion criteria: human studies, analyzing prediction of induction chemotherapy outcome or toxicity based on the pretreatment (or after the first cycle, if more cycles of induction were administered) data, published after the year 2015. The studies regarding metastatic and recurrent cancers as well as the prognosis of overall survival or the outcome of consecutive treatment were not taken into consideration. As revealed from the systematic inspection of the papers, there are over 100 independent parameters analyzed for their suitability as prognostic markers in HNSCC patients undergoing induction chemotherapy. Some of them are promising, but usually they lack important features such as high specificity and sensitivity, low cost, high positive predictive value, clinical relevance, short turnaround time, etc. Subsequent studies are necessary to confirm the usability of the biomarkers for personal medicine.
Collapse
Affiliation(s)
- Łukasz Boguszewicz
- Department of Medical Physics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Warszawa, Poland
| |
Collapse
|
45
|
Park J, Jia S, Salter D, Bagnaninchi P, Hansen CG. The Hippo pathway drives the cellular response to hydrostatic pressure. EMBO J 2022; 41:e108719. [PMID: 35702882 PMCID: PMC9251841 DOI: 10.15252/embj.2021108719] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Cells need to rapidly and precisely react to multiple mechanical and chemical stimuli in order to ensure precise context-dependent responses. This requires dynamic cellular signalling events that ensure homeostasis and plasticity when needed. A less well-understood process is cellular response to elevated interstitial fluid pressure, where the cell senses and responds to changes in extracellular hydrostatic pressure. Here, using quantitative label-free digital holographic imaging, combined with genome editing, biochemical assays and confocal imaging, we analyse the temporal cellular response to hydrostatic pressure. Upon elevated cyclic hydrostatic pressure, the cell responds by rapid, dramatic and reversible changes in cellular volume. We show that YAP and TAZ, the co-transcriptional regulators of the Hippo signalling pathway, control cell volume and that cells without YAP and TAZ have lower plasma membrane tension. We present direct evidence that YAP/TAZ drive the cellular response to hydrostatic pressure, a process that is at least partly mediated via clathrin-dependent endocytosis. Additionally, upon elevated oscillating hydrostatic pressure, YAP/TAZ are activated and induce TEAD-mediated transcription and expression of cellular components involved in dynamic regulation of cell volume and extracellular matrix. This cellular response confers a feedback loop that allows the cell to robustly respond to changes in interstitial fluid pressure.
Collapse
Affiliation(s)
- Jiwon Park
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Siyang Jia
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Donald Salter
- Centre for Genomic & Experimental MedicineMRC Institute of Genetics & Molecular MedicineThe University of Edinburgh, Western General HospitalEdinburghUK
| | - Pierre Bagnaninchi
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Carsten G Hansen
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| |
Collapse
|
46
|
Tiong KL, Lin YW, Yeang CH. Characterization of gene cluster heterogeneity in single-cell transcriptomic data within and across cancer types. Biol Open 2022; 11:275538. [PMID: 35665803 PMCID: PMC9235070 DOI: 10.1242/bio.059256] [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: 01/27/2022] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Despite the remarkable progress in probing tumor transcriptomic heterogeneity by single-cell RNA sequencing (sc-RNAseq) data, several gaps exist in prior studies. Tumor heterogeneity is frequently mentioned but not quantified. Clustering analyses typically target cells rather than genes, and differential levels of transcriptomic heterogeneity of gene clusters are not characterized. Relations between gene clusters inferred from multiple datasets remain less explored. We provided a series of quantitative methods to analyze cancer sc-RNAseq data. First, we proposed two quantitative measures to assess intra-tumoral heterogeneity/homogeneity. Second, we established a hierarchy of gene clusters from sc-RNAseq data, devised an algorithm to reduce the gene cluster hierarchy to a compact structure, and characterized the gene clusters with functional enrichment and heterogeneity. Third, we developed an algorithm to align the gene cluster hierarchies from multiple datasets to a small number of meta gene clusters. By applying these methods to nine cancer sc-RNAseq datasets, we discovered that cancer cell transcriptomes were more homogeneous within tumors than the accompanying normal cells. Furthermore, many gene clusters from the nine datasets were aligned to two large meta gene clusters, which had high and low heterogeneity and were enriched with distinct functions. Finally, we found the homogeneous meta gene cluster retained stronger expression coherence and associations with survival times in bulk level RNAseq data than the heterogeneous meta gene cluster, yet the combinatorial expression patterns of breast cancer subtypes in bulk level data were not preserved in single-cell data. The inference outcomes derived from nine cancer sc-RNAseq datasets provide insights about the contributing factors for transcriptomic heterogeneity of cancer cells and complex relations between bulk level and single-cell RNAseq data. They demonstrate the utility of our methods to enable a comprehensive characterization of co-expressed gene clusters in a wide range of sc-RNAseq data in cancers and beyond. Summary: We propose quantitative methods to analyze cancer sc-RNAseq data: measures of intra-tumoral heterogeneity, characterization of a hierarchy of gene clusters, and alignment of gene cluster hierarchies from multiple datasets.
Collapse
Affiliation(s)
- Khong-Loon Tiong
- Institute of Statistical Science, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| | - Yu-Wei Lin
- Institute of Statistical Science, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan.,The University of Texas MD Anderson Cancer Center, School of Health Profession, Master Program of Diagnostic Genetics, Houston, Texas, 77030, USA
| | - Chen-Hsiang Yeang
- Institute of Statistical Science, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| |
Collapse
|
47
|
Liu Z, Parida S, Wu S, Sears CL, Sharma D, Barman I. Label-Free Vibrational and Quantitative Phase Microscopy Reveals Remarkable Pathogen-Induced Morphomolecular Divergence in Tumor-Derived Cells. ACS Sens 2022; 7:1495-1505. [PMID: 35583030 DOI: 10.1021/acssensors.2c00232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Delineating the molecular and morphological changes that cancer cells undergo in response to extracellular stimuli is crucial for identifying factors that promote tumor progression. Label-free optical imaging offers a potentially promising route for retrieving such single-cell information by generating detailed visualization of the morphology and determining alterations in biomolecular composition. The potential of such nonperturbative morphomolecular microscopy for analyzing microbiota-cancer cell interactions has been surprisingly underappreciated, despite the growing evidence of the critical role of dysbiosis in malignant transformations. Here, using a model system of breast cancer cells, we show that label-free Raman microspectroscopy and quantitative phase microscopy can detect biomolecular and morphological changes in single cells exposed to Bacteroides fragilis toxin (BFT), a toxin secreted by enterotoxigenicB. fragilis. Remarkably, using machine learning to elucidate subtle, but consistent, cellular differences, we found that the morphomolecular differences between BFT-exposed and control breast cancer cells became more accentuated after in vivo passage, corroborating our findings that a short-term BFT exposure imparts a long-term effect on cancer cells and promotes a more invasive phenotype. Complementing more classical labeling techniques, our label-free platform offers a global detection approach with measurements representative of the overall cellular phenotype, paving the way for further investigations into the multifaceted interactions between the cancer cell and the microbiota.
Collapse
Affiliation(s)
- Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Sheetal Parida
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Shaoguang Wu
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Cynthia L. Sears
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
48
|
Shendy NAM, Zimmerman MW, Abraham BJ, Durbin AD. Intrinsic transcriptional heterogeneity in neuroblastoma guides mechanistic and therapeutic insights. Cell Rep Med 2022; 3:100632. [PMID: 35584622 PMCID: PMC9133465 DOI: 10.1016/j.xcrm.2022.100632] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/24/2022] [Accepted: 04/20/2022] [Indexed: 12/20/2022]
Abstract
Cell state is controlled by master transcription factors (mTFs) that determine the cellular gene expression program. Cancer cells acquire dysregulated gene expression programs by mutational and non-mutational processes. Intratumoral heterogeneity can result from cells displaying distinct mTF-regulated cell states, which co-exist within the tumor. One archetypal tumor associated with transcriptionally regulated heterogeneity is high-risk neuroblastoma (NB). Patients with NB have poor overall survival despite intensive therapies, and relapsed patients are commonly refractory to treatment. The cellular populations that comprise NB are marked by different cohorts of mTFs and differential sensitivity to conventional therapies. Recent studies have highlighted mechanisms by which NB cells dynamically shift the cell state with treatment, revealing new opportunities to control the cellular response to treatment by manipulating cell-state-defining transcriptional programs. Here, we review recent advances in understanding transcriptionally defined cancer heterogeneity. We offer challenges to the field to encourage translation of basic science into clinical benefit.
Collapse
Affiliation(s)
- Noha A M Shendy
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mark W Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Adam D Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
49
|
Han Y, Wang D, Peng L, Huang T, He X, Wang J, Ou C. Single-cell sequencing: a promising approach for uncovering the mechanisms of tumor metastasis. J Hematol Oncol 2022; 15:59. [PMID: 35549970 PMCID: PMC9096771 DOI: 10.1186/s13045-022-01280-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
Single-cell sequencing (SCS) is an emerging high-throughput technology that can be used to study the genomics, transcriptomics, and epigenetics at a single cell level. SCS is widely used in the diagnosis and treatment of various diseases, including cancer. Over the years, SCS has gradually become an effective clinical tool for the exploration of tumor metastasis mechanisms and the development of treatment strategies. Currently, SCS can be used not only to analyze metastasis-related malignant biological characteristics, such as tumor heterogeneity, drug resistance, and microenvironment, but also to construct metastasis-related cell maps for predicting and monitoring the dynamics of metastasis. SCS is also used to identify therapeutic targets related to metastasis as it provides insights into the distribution of tumor cell subsets and gene expression differences between primary and metastatic tumors. Additionally, SCS techniques in combination with artificial intelligence (AI) are used in liquid biopsy to identify circulating tumor cells (CTCs), thereby providing a novel strategy for treating tumor metastasis. In this review, we summarize the potential applications of SCS in the field of tumor metastasis and discuss the prospects and limitations of SCS to provide a theoretical basis for finding therapeutic targets and mechanisms of metastasis.
Collapse
Affiliation(s)
- Yingying Han
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lushan Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Tao Huang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, 410031, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
50
|
LeBleu VS, Thiery JP. The Continuing Search for Causality between Epithelial-to-Mesenchymal Transition and the Metastatic Fitness of Carcinoma Cells. Cancer Res 2022; 82:1467-1469. [DOI: 10.1158/0008-5472.can-22-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022]
Abstract
Abstract
The epithelial-to-mesenchymal transition (EMT) is an epithelial plasticity program that is associated with embryonic development and organogenesis, and which resurfaces to a certain extent following epithelial injury caused by inflammation, fibrosis, and carcinoma progression. Carcinoma cell EMT plasticity programs superimposed on inherent genetic defects have been implicated as important for metastatic dissemination and secondary tumor formation. A careful review of data-driven facts suggests that a causal relationship between any degree of EMT program and metastasis continues to be elusive, and the steps of metastasis likely involve other mechanisms influenced by the carcinoma cell genotype and the tumor microenvironment.
Collapse
Affiliation(s)
- Valerie S. LeBleu
- Feinberg School of Medicine and Kellogg School of Management, Northwestern University, Chicago, Illinois
| | - Jean Paul Thiery
- Guangzhou Laboratory, Guangzhou International Bioisland, Guangzhou, P.R. China
| |
Collapse
|