1
|
Lee E, Hong JJ, Samcam Vargas G, Sauerwald N, Wei Y, Hang X, Theesfeld CL, Volmar JAA, Miller JM, Wang W, Wang S, Laevsky G, DeCoste CJ, Kang Y. CXCR4 + mammary gland macrophageal niche promotes tumor initiating cell activity and immune suppression during tumorigenesis. Nat Commun 2025; 16:4854. [PMID: 40413176 PMCID: PMC12103607 DOI: 10.1038/s41467-025-59972-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Tumor-initiating cells (TICs) share features and regulatory pathways with normal stem cells, yet how the stem cell niche contributes to tumorigenesis remains unclear. Here, we identify CXCR4+ macrophages as a niche population enriched in normal mammary ducts, where they promote the regenerative activity of basal cells in response to luminal cell-derived CXCL12. CXCL12 triggers AKT-mediated stabilization of β-catenin, which induces Wnt ligands and pro-migratory genes, enabling intraductal macrophage infiltration and supporting regenerative activity of basal cells. Notably, these same CXCR4+ niche macrophages regulate the tumor-initiating activity of various breast cancer subtypes by enhancing TIC survival and tumor-forming capacity, while promoting early immune evasion through regulatory T cell induction. Furthermore, a CXCR4+ niche macrophage gene signature correlates with poor prognosis in human breast cancer. These findings highlight the pivotal role of the CXCL12-CXCR4 axis in orchestrating interactions between niche macrophages, mammary epithelial cells, and immune cells, thereby establishing a supportive niche for both normal tissue regeneration and mammary tumor initiation.
Collapse
Affiliation(s)
- Eunmi Lee
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Jason J Hong
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | | | - Natalie Sauerwald
- Center for Computational Biology, Flatiron Institute, New York, NY, 10010, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Xiang Hang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Chandra L Theesfeld
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Jean Arly A Volmar
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Jennifer M Miller
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Wei Wang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Sha Wang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Gary Laevsky
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Christina J DeCoste
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
2
|
Merle C, Rodrigues C, Pourkhalili Langeroudi A, Journot R, Rost F, Dang Y, Rulands S, Fre S. Transcriptional landscapes underlying Notch-induced lineage conversion and plasticity of mammary basal cells. EMBO J 2025; 44:2827-2855. [PMID: 40186028 DOI: 10.1038/s44318-025-00424-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
The mammary epithelium derives from multipotent mammary stem cells (MaSCs) that engage into differentiation during embryonic development. However, adult MaSCs maintain the ability to reactivate multipotency in non-physiological contexts. We previously reported that Notch1 activation in committed basal cells triggers a basal-to-luminal cell fate switch in the mouse mammary gland. Here, we report conservation of this mechanism and found that in addition to the mammary gland, constitutive Notch1 signaling induces a basal-to-luminal cell fate switch in adult cells of the lacrimal gland, the salivary gland, and the prostate. Since the lineage transition is progressive in time, we performed single-cell transcriptomic analysis on index-sorted mammary cells at different stages of lineage conversion, generating a temporal map of changes in cell identity. Combining single-cell analyses with organoid assays, we demonstrate that cell proliferation is indispensable for this lineage conversion. We also reveal the individual transcriptional landscapes underlying the cellular plasticity switching of committed mammary cells in vivo with spatial and temporal resolution. Given the roles of Notch signaling in cancer, these results may help to better understand the mechanisms that drive cellular transformation.
Collapse
Affiliation(s)
- Candice Merle
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Calvin Rodrigues
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Atefeh Pourkhalili Langeroudi
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Robin Journot
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Fabian Rost
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Yiteng Dang
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
| | - Steffen Rulands
- Ludwig-Maximilians-Universität München, Arnold-Sommerfeld-Center for Theoretical Physics, München, Germany
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France.
| |
Collapse
|
3
|
Bui H, Andersson S, Sola-Carvajal A, De Marchi T, Vähäkangas E, Holopainen M, House AH, Rovenko BM, Englund JI, Kasper M, Kuuluvainen E, Käkelä R, Hietakangas V, Niméus E, Katajisto P. Glucose-6-phosphate-dehydrogenase on old peroxisomes maintains self-renewal of epithelial stem cells after asymmetric cell division. Nat Commun 2025; 16:3932. [PMID: 40287409 PMCID: PMC12033372 DOI: 10.1038/s41467-025-58752-z] [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: 11/06/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Selective inheritance of sub-cellular components has emerged as a mechanism guiding stem cell fate after asymmetric cell divisions. Peroxisomes play a crucial role in multiple metabolic processes such as fatty acid metabolism and reactive oxygen species detoxification, but the apportioning of peroxisomes during stem cell division remains understudied. Here, we develop a mouse model and labeling technique to follow the dynamics of distinct peroxisome age-classes, and find that old peroxisomes are inherited by the daughter cell retaining full stem cell potency in mammary and epidermal stem cell divisions. Old peroxisomes carry Glucose-6-phosphate-dehydrogenase, whose specific location on the peroxisomal membrane promotes stem cell function by facilitating peroxisomal ether lipid synthesis. Our study demonstrates age-selective apportioning of peroxisomes in vivo, and unveils how functional heterogeneity of peroxisomes is utilized by asymmetrically dividing cells to metabolically divert the fate of the two daughter cells.
Collapse
Grants
- ERC, #677809, and #101045009 EC | EC Seventh Framework Programm | FP7 Ideas: European Research Council (FP7-IDEAS-ERC - Specific Programme: "Ideas" Implementing the Seventh Framework Programme of the European Community for Research, Technological Development and Demonstration Activities (2007 to 2013))
- #266869, #304591, #312436, #320185 Academy of Finland (Suomen Akatemia)
- 2018-03078, 2018-02963, 2022-01304 Vetenskapsrådet (Swedish Research Council)
- 190634, 180681, and 222499 Cancerfonden (Swedish Cancer Society)
- KAW 2014.0207 and 20220054 Knut och Alice Wallenbergs Stiftelse (Knut and Alice Wallenberg Foundation)
- Syöpäjärjestöt (Cancer Society of Finland)
- Chan Zuckerberg Initiative MET-0000000418 Center for Innovative Medicine CIMED Sigrid Juselius Foundation
- Finnish Cultural Foundation | Uudenmaan Rahasto (Uusimaa Regional Fund)
- Maud Kuistilan Muistosäätiö (Maud Kuistila Memorial Foundation)
- Doctoral Programme in Biomedicine at the University of Helsinki
Collapse
Affiliation(s)
- Hien Bui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Simon Andersson
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Agustin Sola-Carvajal
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Tommaso De Marchi
- Division of Oncology and Surgery, Department of Clinical Sciences, Lund University, 22362, Lund, Sweden
| | - Eliisa Vähäkangas
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
- Stem cells and metabolism research program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Minna Holopainen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE), and Biocenter Finland, University of Helsinki, 00014, Helsinki, Finland
| | - Andrew H House
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE), and Biocenter Finland, University of Helsinki, 00014, Helsinki, Finland
| | - Bohdana M Rovenko
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Johanna I Englund
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Emilia Kuuluvainen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE), and Biocenter Finland, University of Helsinki, 00014, Helsinki, Finland
| | - Ville Hietakangas
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland
| | - Emma Niméus
- Division of Oncology and Surgery, Department of Clinical Sciences, Lund University, 22362, Lund, Sweden
- Department of Surgery, Skåne University Hospital, 22242, Lund, Sweden
| | - Pekka Katajisto
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790, Helsinki, Finland.
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790, Helsinki, Finland.
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| |
Collapse
|
4
|
Si H, Mendoza Mendoza E, Esquivel M, Creighton CJ, Xu J, Roarty K. Noncanonical Wnt/Ror2 Signaling Regulates Basal Cell Fidelity and Branching Morphogenesis in the Mammary Gland. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640099. [PMID: 40060578 PMCID: PMC11888327 DOI: 10.1101/2025.02.25.640099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The mammary gland epithelium relies on a delicate balance between basal and luminal cell lineages to maintain tissue homeostasis and enable proper development. While the role of canonical Wnt signaling in mammary biology is well-established, the contribution of noncanonical Wnt signaling to lineage identity has remained unclear. Noncanonical Wnt pathways are primarily associated with morphogenesis, cytoskeletal regulation, and cell migration, but whether they are required for maintaining epithelial cell fate remains largely unexplored. Here, we demonstrate that the noncanonical Wnt receptor Ror2 is expressed in both basal and luminal lineages, yet selectively maintained in basal cells throughout development, suggesting a lineage-specific function. Using a p63CreERT2/+ lineage-specific mouse model, we show that Ror2 deletion in basal epithelial cells enhances secondary and tertiary branching while driving a basal-to-luminal fate transition, marked by downregulation of basal markers (K14, K5) and upregulation of luminal markers (K8, K18, ERα). Mechanistically, Ror2 loss disrupts RhoA-ROCK1-YAP1 signaling, leading to cytoskeletal reorganization, chromatin remodeling, and increased accessibility at luminal regulatory loci. Notably, ROCK1 inhibition phenocopies Ror2 loss, reinforcing the critical role of the RhoA-ROCK1 axis in basal cell maintenance. These findings provide direct genetic and mechanistic evidence that noncanonical Wnt signaling is essential for maintaining basal lineage fidelity, offering new insights into the mechanisms regulating epithelial plasticity. Given the fundamental importance of lineage stability in epithelial homeostasis, our results suggest that disruptions in Wnt/Ror2 signaling may contribute to aberrant fate transitions relevant to breast cancer progression.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Erika Mendoza Mendoza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Madelyn Esquivel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Chad J. Creighton
- Dan L. Duncan Comprehensive Cancer Center, Breast Cancer Program, Baylor College of Medicine, Houston, TX 77030
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Comprehensive Cancer Center, Breast Cancer Program, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
5
|
Worley J, Noh H, You D, Turunen MM, Ding H, Paull E, Griffin AT, Grunn A, Zhang M, Guillan K, Bush EC, Brosius SJ, Hibshoosh H, Mundi PS, Sims P, Dalerba P, Dela Cruz FS, Kung AL, Califano A. Identification and Pharmacological Targeting of Treatment-Resistant, Stem-like Breast Cancer Cells for Combination Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.08.562798. [PMID: 38798673 PMCID: PMC11118419 DOI: 10.1101/2023.11.08.562798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Tumors frequently harbor isogenic yet epigenetically distinct subpopulations of multi-potent cells with high tumor-initiating potential-often called Cancer Stem-Like Cells (CSLCs). These can display preferential resistance to standard-of-care chemotherapy. Single-cell analyses can help elucidate Master Regulator (MR) proteins responsible for governing the transcriptional state of these cells, thus revealing complementary dependencies that may be leveraged via combination therapy. Interrogation of single-cell RNA sequencing profiles from seven metastatic breast cancer patients, using perturbational profiles of clinically relevant drugs, identified drugs predicted to invert the activity of MR proteins governing the transcriptional state of chemoresistant CSLCs, which were then validated by CROP-seq assays. The top drug, the anthelmintic albendazole, depleted this subpopulation in vivo without noticeable cytotoxicity. Moreover, sequential cycles of albendazole and paclitaxel-a commonly used chemotherapeutic -displayed significant synergy in a patient-derived xenograft (PDX) from a TNBC patient, suggesting that network-based approaches can help develop mechanism-based combinatorial therapies targeting complementary subpopulations. Statement of significance Network-based approaches, as shown in a study on metastatic breast cancer, can develop effective combinatorial therapies targeting complementary subpopulations. By analyzing scRNA-seq data and using clinically relevant drugs, researchers identified and depleted chemoresistant Cancer Stem-Like Cells, enhancing the efficacy of standard chemotherapies.
Collapse
Affiliation(s)
- Jeremy Worley
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Heeju Noh
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mikko M Turunen
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Hongxu Ding
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pharmacy Practice & Science, College of Pharmacy, University of Arizona, Tucson, Arizona, USA 85721
| | - Evan Paull
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Aaron T Griffin
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Adina Grunn
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Mingxuan Zhang
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Kristina Guillan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Erin C Bush
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Samantha J Brosius
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, USA 10032
| | - Prabhjot S Mundi
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
| | - Peter Sims
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Piero Dalerba
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, USA 10032
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, USA 10032
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Califano
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Biochemistry & Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY USA 10032
| |
Collapse
|
6
|
Hakala S, Hämäläinen A, Sandelin S, Giannareas N, Närvä E. Detection of Cancer Stem Cells from Patient Samples. Cells 2025; 14:148. [PMID: 39851576 PMCID: PMC11764358 DOI: 10.3390/cells14020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Närvä
- Institute of Biomedicine and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (S.H.); (A.H.); (S.S.); (N.G.)
| |
Collapse
|
7
|
Filippi A, Deculescu-Ioniță T, Hudiță A, Baldasici O, Gălățeanu B, Mocanu MM. Molecular Mechanisms of Dietary Compounds in Cancer Stem Cells from Solid Tumors: Insights into Colorectal, Breast, and Prostate Cancer. Int J Mol Sci 2025; 26:631. [PMID: 39859345 PMCID: PMC11766403 DOI: 10.3390/ijms26020631] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer stem cells (CSC) are known to be the main source of tumor relapse, metastasis, or multidrug resistance and the mechanisms to counteract or eradicate them and their activity remain elusive. There are different hypotheses that claim that the origin of CSC might be in regular stem cells (SC) and, due to accumulation of mutations, these normal cells become malignant, or the source of CSC might be in any malignant cell that, under certain environmental circumstances, acquires all the qualities to become CSC. Multiple studies indicate that lifestyle and diet might represent a source of wellbeing that can prevent and ameliorate the malignant phenotype of CSC. In this review, after a brief introduction to SC and CSC, we analyze the effects of phenolic and non-phenolic dietary compounds and we highlight the molecular mechanisms that are shown to link diets to CSC activation in colon, breast, and prostate cancer. We focus the analysis on specific markers such as sphere formation, CD surface markers, epithelial-mesenchymal transition (EMT), Oct4, Nanog, Sox2, and aldehyde dehydrogenase 1 (ALDH1) and on the major signaling pathways such as PI3K/Akt/mTOR, NF-κB, Notch, Hedgehog, and Wnt/β-catenin in CSC. In conclusion, a better understanding of how bioactive compounds in our diets influence the dynamics of CSC can raise valuable awareness towards reducing cancer risk.
Collapse
Affiliation(s)
- Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Teodora Deculescu-Ioniță
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Oana Baldasici
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Maria-Magdalena Mocanu
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| |
Collapse
|
8
|
Van Keymeulen A. Mechanisms of Regulation of Cell Fate in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:167-184. [PMID: 39821026 DOI: 10.1007/978-3-031-70875-6_10] [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: 01/19/2025]
Abstract
This chapter focuses on the mechanisms of regulation of cell fate in breast development, occurring mainly after birth, as well as in breast cancer. First, we will review how the microenvironment of the breast, as well as external cues, plays a crucial role in mammary gland cell specification and will describe how it has been shown to reprogram non-mammary cells into mammary epithelial cells. Then we will focus on the transcription factors and master regulators which have been established to be determinant for basal (BC) and luminal cell (LC) identity, and will describe the experiments of ectopic expression or loss of function of these transcription factors which demonstrated that they were crucial for cell fate. We will also discuss how master regulators are involved in the fate choice of LCs between estrogen receptor (ER)-positive cells and ER- cells, which will give rise to alveolar cells upon pregnancy and lactation. We will describe how oncogene expression induces reprogramming and change of fate of mammary epithelial cells before tumor appearance, which could be an essential step in tumorigenesis. Finally, we will describe the involvement of master regulators of mammary epithelial cells in breast cancer.
Collapse
Affiliation(s)
- Alexandra Van Keymeulen
- Laboratory of Stem Cells and Cancer (LSCC), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
9
|
Abatti LE, Gillespie ZE, Lado-Fernández P, Collado M, Mitchell JA. A role for NFIB in SOX2 downregulation and epigenome accessibility changes due to long-term estrogen treatment of breast cancer epithelial cells. Biochem Cell Biol 2025; 103:1-14. [PMID: 40009831 DOI: 10.1139/bcb-2024-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025] Open
Abstract
Estrogen (E2) regulates the differentiation and proliferation of mammary progenitor cells by modulating the transcription of multiple genes. One of the genes that is downregulated by E2 is SOX2, a transcription factor associated with stem and progenitor cells that is overexpressed during breast tumourigenesis. To elucidate the mechanisms underlying E2-mediated SOX2 repression, we investigated epigenome and transcriptome changes following short- and long-term E2 exposure in breast cancer cells. We found that short-term E2 exposure reduces chromatin accessibility at the downstream SOX2 SRR134 enhancer, decreasing SOX2 expression. In contrast, long-term E2 exposure completely represses SOX2 transcription while maintaining accessibility at the SRR124-134 enhancer cluster, keeping it poised for reactivation. This repression was accompanied by widespread epigenome and transcriptome changes associated with commitment towards a more differentiated and less invasive luminal phenotype. Finally, we identified a role for the transcription factor NFIB in this process, suggesting it collaborates with the estrogen receptor to mediate SOX2 repression and genome-wide epigenome accessibility changes.
Collapse
Affiliation(s)
- Luis E Abatti
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Zoe E Gillespie
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Patricia Lado-Fernández
- Laboratory of Cell Senescence, Cancer and Aging, Health Research Institute of Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Collado
- Laboratory of Cell Senescence, Cancer and Aging, Health Research Institute of Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Iggo R, MacGrogan G. Classification of Breast Cancer Through the Perspective of Cell Identity Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:185-207. [PMID: 39821027 DOI: 10.1007/978-3-031-70875-6_11] [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: 01/19/2025]
Abstract
The mammary epithelium has an inner luminal layer that contains estrogen receptor (ER)-positive hormone-sensing cells and ER-negative alveolar/secretory cells, and an outer basal layer that contains myoepithelial/stem cells. Most human tumours resemble either hormone-sensing cells or alveolar/secretory cells. The most widely used molecular classification, the Intrinsic classification, assigns hormone-sensing tumours to Luminal A/B and human epidermal growth factor 2-enriched (HER2E)/molecular apocrine (MA)/luminal androgen receptor (LAR)-positive classes, and alveolar/secretory tumours to the Basal-like class. Molecular classification is most useful when tumours have classic invasive carcinoma of no special type (NST) histology. It is less useful for special histological types of breast cancer, such as metaplastic breast cancer and adenoid cystic cancer, which are better described with standard pathology terms. Compared to mice, humans show a strong bias towards making tumours that resemble mammary hormone-sensing cells. This could be caused by the formation in adolescence of der(1;16), a translocation through the centromeres of chromosomes 1 and 16, which only occurs in humans and could trap the cells in the hormone-sensing state.
Collapse
Affiliation(s)
- Richard Iggo
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France.
| | - Gaetan MacGrogan
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France
| |
Collapse
|
11
|
Clarke RB, Sørlie T. A Guide to Breast Cancer Research: An Introduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:1-5. [PMID: 39821017 DOI: 10.1007/978-3-031-70875-6_1] [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: 01/19/2025]
Abstract
"A Guide to Breast Cancer Research: From Cells and Molecular Mechanisms to Therapy" is designed as a comprehensive reference for early career investigators and postgraduate students. This book aims to provide a broad overview of contemporary breast cancer research. It covers key areas including development and cancer, metastasis and immunology, subtypes, signalling, therapy, and resistance. This book is organised into seven sections addressing mammary gland development, model systems, cellular origins and heterogeneity, cellular and molecular bases, signalling pathways, metastasis and immunity, and treatment and resistance mechanisms. A few topics such as specific signalling pathways, some emerging therapies, imaging technologies, and AI applications are only briefly mentioned or omitted, reflecting the ever-evolving nature of breast cancer research. This book emphasises the importance of collaboration in advancing cancer research. Initiatives like the Cancer Moonshot and Cancer Grand Challenges advocate for "radical collaboration" of researchers with shared visions and resources. We also note the significance of global efforts in breast cancer research, the need for addressing disparities in care across different regions and for equity in healthcare. Overall, this book showcases milestones and advances in breast cancer research over the past three decades, reflecting significant progress in understanding and treating the disease, which has led to improved patient outcomes.
Collapse
Affiliation(s)
- Robert B Clarke
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, Manchester Breast Centre, University of Manchester, Manchester, UK
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
12
|
Jiang C, Centonze A, Song Y, Chrisnandy A, Tika E, Rezakhani S, Zahedi Z, Bouvencourt G, Dubois C, Van Keymeulen A, Lütolf M, Sifrim A, Blanpain C. Collagen signaling and matrix stiffness regulate multipotency in glandular epithelial stem cells in mice. Nat Commun 2024; 15:10482. [PMID: 39695111 PMCID: PMC11655882 DOI: 10.1038/s41467-024-54843-5] [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: 07/28/2023] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Glandular epithelia, including mammary gland (MG) and prostate, are composed of luminal and basal cells. During embryonic development, glandular epithelia arise from multipotent stem cells (SCs) that are replaced after birth by unipotent basal and unipotent luminal SCs. Different conditions, such as basal cell transplantation, luminal cell ablation, and oncogene expression can reinduce adult basal SC (BaSCs) multipotency in different glandular epithelia. The mechanisms regulating the reactivation of multipotency are incompletely understood. Here, we have found that Collagen I expression is commonly upregulated in BaSCs across the different multipotent conditions. Increasing collagen concentration or stiffness of the extracellular matrix (ECM) promotes BaSC multipotency in MG and prostate organoids. Single cell RNA-seq of MG organoids in stiff conditions have uncovered the importance of β1 integrin/FAK/AP-1 axis in the regulation of BaSC multipotency. Altogether our study uncovers the key role of Collagen signaling and ECM stiffness in the regulation of multipotency in glandular epithelia.
Collapse
Affiliation(s)
- Chen Jiang
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alessia Centonze
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yura Song
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Antonius Chrisnandy
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Elisavet Tika
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Saba Rezakhani
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zahra Zahedi
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Gaëlle Bouvencourt
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christine Dubois
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Matthias Lütolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL, Lausanne, Switzerland
- Institute of Human Biology (IHB), Pharma Research and Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alejandro Sifrim
- Laboratory of Multi-Omic Integrative Bioinformatics (LMIB), Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- WEL Research Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
13
|
Williams MJ, Oliphant MUJ, Au V, Liu C, Baril C, O'Flanagan C, Lai D, Beatty S, Van Vliet M, Yiu JC, O'Connor L, Goh WL, Pollaci A, Weiner AC, Grewal D, McPherson A, Norton K, Moore M, Prabhakar V, Agarwal S, Garber JE, Dillon DA, Shah SP, Brugge JS, Aparicio S. Luminal breast epithelial cells of BRCA1 or BRCA2 mutation carriers and noncarriers harbor common breast cancer copy number alterations. Nat Genet 2024; 56:2753-2762. [PMID: 39567747 PMCID: PMC11631757 DOI: 10.1038/s41588-024-01988-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
The prevalence and nature of somatic copy number alterations (CNAs) in breast epithelium and their role in tumor initiation and evolution remain poorly understood. Using single-cell DNA sequencing (49,238 cells) of epithelium from BRCA1 and BRCA2 carriers or wild-type individuals, we identified recurrent CNAs (for example, 1q-gain and 7q, 10q, 16q and 22q-loss) that are present in a rare population of cells across almost all samples (n = 28). In BRCA1/BRCA2 carriers, these occur before loss of heterozygosity (LOH) of wild-type alleles. These CNAs, common in malignant tumors, are enriched in luminal cells but absent in basal myoepithelial cells. Allele-specific analysis of prevalent CNAs reveals that they arose by independent mutational events, consistent with convergent evolution. BRCA1/BRCA2 carriers contained a small percentage of cells with extreme aneuploidy, featuring loss of TP53, BRCA1/BRCA2 LOH and multiple breast cancer-associated CNAs. Our findings suggest that CNAs arising in normal luminal breast epithelium are precursors to clonally expanded tumor genomes.
Collapse
Affiliation(s)
- Marc J Williams
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael U J Oliphant
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School (HMS), Boston, MA, USA
| | - Vinci Au
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cathy Liu
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Caroline Baril
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ciara O'Flanagan
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Lai
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sean Beatty
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Van Vliet
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jacky Ch Yiu
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lauren O'Connor
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School (HMS), Boston, MA, USA
| | - Walter L Goh
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School (HMS), Boston, MA, USA
| | - Alicia Pollaci
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Adam C Weiner
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diljot Grewal
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andrew McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Klarisa Norton
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School (HMS), Boston, MA, USA
| | - McKenna Moore
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Vikas Prabhakar
- Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA, USA
| | - Shailesh Agarwal
- Department of Surgery, Brigham and Women's Hospital (BWH), Boston, MA, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Deborah A Dillon
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Joan S Brugge
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School (HMS), Boston, MA, USA.
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
14
|
Xie Y, Liu Z, Zhang J, Li G, Ni B, Shi C, Zou Y, Zhou Y, Shang X. Deciphering the composition and key driver genes of breast invasive micropapillary carcinoma by multi-omics analysis. iScience 2024; 27:111178. [PMID: 39524324 PMCID: PMC11549989 DOI: 10.1016/j.isci.2024.111178] [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: 04/07/2024] [Revised: 08/22/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
In this study, we delved into the intrinsic cellular components and transcriptomic signatures characterizing breast-invasive micropapillary carcinoma (IMPC). Employing bulk RNA sequencing, we conducted differential gene expression and functional profiles across breast cancer tissues. Single-cell transcriptome sequencing was performed on mixed IMPC samples. Moreover, a multicenter retrospective cohort of IMPC patients validated the critical role of KRT80. Our findings illuminated heightened activity in redox reactions and metabolism-related functions within IMPC compared to other tissue types. The single-cell atlas of IMPC demonstrated substantial heterogeneity predominantly driven by two distinct cell subsets: epithelioid and interstitial cells. Pseudotime analysis unveiled unique cell trajectories, and we found positive correlation between KRT80 expression and clinicopathological characteristics in IMPC. High KRT80 expression was associated with shorter overall survival for IMPC patients. This investigation unmasked extensive heterogeneity within breast IMPC tumors, delineating lineage distinctions across diverse cell clusters. It unveils potential prospective therapeutic targets with clinical relevance.
Collapse
Affiliation(s)
- Yongjie Xie
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ziyun Liu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jie Zhang
- Tianjin Medical University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Guangming Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bo Ni
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chunlei Shi
- Department of colorectal Surgery, The Wuhu Hospital of Traditional Chinese Medicine, Anhui, China
| | - Yiping Zou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yaoyao Zhou
- Tianjin Medical University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaobin Shang
- Department of Minimally Invasive Esophageal Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
15
|
Vallmajo-Martin Q, Ma Z, Srinivasan S, Murali D, Dravis C, Mukund K, Subramaniam S, Wahl GM, Lytle NK. The molecular chronology of mammary epithelial cell fate switching. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617155. [PMID: 39415993 PMCID: PMC11482796 DOI: 10.1101/2024.10.08.617155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The adult mammary gland is maintained by lineage-restricted progenitor cells through pregnancy, lactation, involution, and menopause. Injury resolution, transplantation-associated mammary gland reconstitution, and tumorigenesis are unique exceptions, wherein mammary basal cells gain the ability to reprogram to a luminal state. Here, we leverage newly developed cell-identity reporter mouse strains, and time-resolved single-cell epigenetic and transcriptomic analyses to decipher the molecular programs underlying basal-to-luminal fate switching in vivo. We demonstrate that basal cells rapidly reprogram toward plastic cycling intermediates that appear to hijack molecular programs we find in bipotent fetal mammary stem cells and puberty-associatiated cap cells. Loss of basal-cell specifiers early in dedifferentiation coincides with activation of Notch and BMP, among others. Pharmacologic blockade of each pathway disrupts basal-to-luminal transdifferentiation. Our studies provide a comprehensive map and resource for understanding the coordinated molecular changes enabling terminally differentiated epithelial cells to transition between cell lineages and highlights the stunning rapidity by which epigenetic reprogramming can occur in response to disruption of tissue structure.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Zhibo Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- These authors contributed equally
| | - Sumana Srinivasan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Divya Murali
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Christopher Dravis
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Geoffrey M. Wahl
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nikki K. Lytle
- Department of Surgery, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- These authors contributed equally
| |
Collapse
|
16
|
Ye Z, Xu Y, Zhang M, Cai C. Sympathetic nerve signals: orchestrators of mammary development and stem cell vitality. J Mol Cell Biol 2024; 16:mjae020. [PMID: 38740522 PMCID: PMC11520406 DOI: 10.1093/jmcb/mjae020] [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: 11/11/2023] [Revised: 03/25/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024] Open
Abstract
The mammary gland is a dynamic organ that undergoes significant changes at multiple stages of postnatal development. Although the roles of systemic hormones and microenvironmental cues in mammary homeostasis have been extensively studied, the influence of neural signals, particularly those from the sympathetic nervous system, remains poorly understood. Here, using a mouse mammary gland model, we delved into the regulatory role of sympathetic nervous signaling in the context of mammary stem cells and mammary development. Our findings revealed that depletion of sympathetic nerve signals results in defective mammary development during puberty, adulthood, and pregnancy, accompanied by a reduction in mammary stem cell numbers. Through in vitro three-dimensional culture and in vivo transplantation analyses, we demonstrated that the absence of sympathetic nerve signals hinders mammary stem cell self-renewal and regeneration, while activation of sympathetic nervous signaling promotes these capacities. Mechanistically, sympathetic nerve signals orchestrate mammary stem cell activity and mammary development through the extracellular signal-regulated kinase signaling pathway. Collectively, our study unveils the crucial roles of sympathetic nerve signals in sustaining mammary development and regulating mammary stem cell activity, offering a novel perspective on the involvement of the nervous system in modulating adult stem cell function and organ development.
Collapse
Affiliation(s)
- Zi Ye
- Department of Pulmonary Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Yu Xu
- Department of Pulmonary Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Mengna Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Cheguo Cai
- Department of Pulmonary Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
17
|
Finot L, Hue-Beauvais C, Aujean E, Le Provost F, Chanat E. Sorted stem/progenitor epithelial cells of pubertal bovine mammary gland present limited potential to reconstitute an organised mammary epithelium after transplantation. PLoS One 2024; 19:e0296614. [PMID: 39423190 PMCID: PMC11488748 DOI: 10.1371/journal.pone.0296614] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 09/29/2024] [Indexed: 10/21/2024] Open
Abstract
The development and maintenance of mammary gland tissue depend on the proliferation and differentiation of mammary stem and progenitor cells. Here, we investigated populations of mammary epithelial cells that are potential candidates for bovine mammary gland development using xenotransplantation into mice cleared mammary fat pad. Transplanted mammary explants from 17-month-old Holstein heifers developed outgrowths exhibiting the archetypal morphology and molecular marker distributions of the bovine gland. Xenotransplantation of sorted mammary epithelial cells (CD49fpos) into bovinised fat pads using inactivated bovine fibroblasts resulted in outgrowth developments with 50% take rate, but these lacked the ductal or alveolar epithelial structures of the normal mammary gland. Similar results were obtained with xenografts of candidate bovine mammary epithelial stem cells (CD49fhighCD24pos) or epithelial cells of the basal lineage (CD49fhighCD24neg) which also developed as clumps of cells surrounded by stromal stretches within the mouse adipose tissue. In conclusion, sorted cells showed compromised regenerative potential for epithelial morphogenesis. Further work is therefore needed to identify mammary stem/progenitor cells with full regenerative capabilities for biogenesis of normal mammary gland structure, with milk-secreting function.
Collapse
Affiliation(s)
- Laurence Finot
- PEGASE, INRAE, Institut Agro, Saint Gilles, Paris, France
| | - Cathy Hue-Beauvais
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Etienne Aujean
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Fabienne Le Provost
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Eric Chanat
- PEGASE, INRAE, Institut Agro, Saint Gilles, Paris, France
| |
Collapse
|
18
|
Feng R, Zhao J, Zhang Q, Zhu Z, Zhang J, Liu C, Zheng X, Wang F, Su J, Ma X, Mi X, Guo L, Yan X, Liu Y, Li H, Chen X, Deng Y, Wang G, Zhang Y, Liu X, Liu J. Generation of Anti-Mastitis Gene-Edited Dairy Goats with Enhancing Lysozyme Expression by Inflammatory Regulatory Sequence using ISDra2-TnpB System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404408. [PMID: 39099401 PMCID: PMC11481229 DOI: 10.1002/advs.202404408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Gene-editing technology has become a transformative tool for the precise manipulation of biological genomes and holds great significance in the field of animal disease-resistant breeding. Mastitis, a prevalent disease in animal husbandry, imposes a substantial economic burden on the global dairy industry. In this study, a regulatory sequence gene editing breeding strategy for the successful creation of a gene-edited dairy (GED) goats with enhanced mastitis resistance using the ISDra2-TnpB system and dairy goats as the model animal is proposed. This included the targeted integration of an innate inflammatory regulatory sequence (IRS) into the promoter region of the lysozyme (LYZ) gene. Upon Escherichia Coli (E. coli) mammary gland infection, GED goats exhibited increased LYZ expression, showing robust anti-mastitis capabilities, mitigating PANoptosis activation, and alleviating blood-milk-barrier (BMB) damage. Notably, LYZ is highly expressed only in E. coli infection. This study marks the advent of anti-mastitis gene-edited animals with exogenous-free gene expression and demonstrates the feasibility of the gene-editing strategy proposed in this study. In addition, it provides a novel gene-editing blueprint for developing disease-resistant strains, focusing on disease specificity and biosafety while providing a research basis for the widespread application of the ISDra2-TnpB system.
Collapse
Affiliation(s)
- Rui Feng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jianglin Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Qian Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Zhenliang Zhu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Junyu Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Chengyuan Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoman Zheng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Fan Wang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jie Su
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xianghai Ma
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoyu Mi
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Lin Guo
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoxue Yan
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yayi Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Huijia Li
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xu Chen
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yi Deng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Guoyan Wang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xu Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| |
Collapse
|
19
|
Chen W, Guo L, Wei W, Cai C, Wu G. Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation is essential for maintenance of mammary stem cell activity. Cell Rep 2024; 43:114762. [PMID: 39321020 DOI: 10.1016/j.celrep.2024.114762] [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: 05/30/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Adult mammary stem cells (aMaSCs) are vital to tissue expansion and remodeling during the process of postnatal mammary development. The protein C receptor (Procr) is one of the well-identified surface markers of multipotent aMaSCs. However, an understanding of the regulatory mechanisms governing Procr's protein stability remains incomplete. In this study, we identified Glycoprotein m6a (Gpm6a) as a critical protein for aMaSC activity modulation by using the Gpm6a knockout mouse model. Interestingly, we determined that Gpm6a depletion results in a reduction of Procr protein stability. Mechanistically, Gpm6a regulates Procr protein stability by mediating the formation of lipid rafts, a process requiring Zdhhc1 and Zdhhc2 to palmitate Gpm6a at Cys17,18 and Cys246 sites. Our findings highlight an important mechanism involving Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation for the regulation of Procr stability, aMaSC activity, and postnatal mammary development.
Collapse
Affiliation(s)
- Weizhen Chen
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Guo
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Wei Wei
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
20
|
Zakic T, Pekovic-Vaughan V, Cvoro A, Korac A, Jankovic A, Korac B. Redox and metabolic reprogramming in breast cancer and cancer-associated adipose tissue. FEBS Lett 2024; 598:2106-2134. [PMID: 38140817 DOI: 10.1002/1873-3468.14794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Redox and metabolic processes are tightly coupled in both physiological and pathological conditions. In cancer, their integration occurs at multiple levels and is characterized by synchronized reprogramming both in the tumor tissue and its specific but heterogeneous microenvironment. In breast cancer, the principal microenvironment is the cancer-associated adipose tissue (CAAT). Understanding how the redox-metabolic reprogramming becomes coordinated in human breast cancer is imperative both for cancer prevention and for the establishment of new therapeutic approaches. This review aims to provide an overview of the current knowledge of the redox profiles and regulation of intermediary metabolism in breast cancer while considering the tumor and CAAT of breast cancer as a unique Warburg's pseudo-organ. As cancer is now recognized as a systemic metabolic disease, we have paid particular attention to the cell-specific redox-metabolic reprogramming and the roles of estrogen receptors and circadian rhythms, as well as their crosstalk in the development, growth, progression, and prognosis of breast cancer.
Collapse
Affiliation(s)
- Tamara Zakic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Vanja Pekovic-Vaughan
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, William Henry Duncan Building, University of Liverpool, UK
| | | | | | - Aleksandra Jankovic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Bato Korac
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Serbia
| |
Collapse
|
21
|
Ciwinska M, Messal HA, Hristova HR, Lutz C, Bornes L, Chalkiadakis T, Harkes R, Langedijk NSM, Hutten SJ, Menezes RX, Jonkers J, Prekovic S, Simons BD, Scheele CLGJ, van Rheenen J. Mechanisms that clear mutations drive field cancerization in mammary tissue. Nature 2024; 633:198-206. [PMID: 39232148 PMCID: PMC11374684 DOI: 10.1038/s41586-024-07882-3] [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: 12/13/2022] [Accepted: 07/26/2024] [Indexed: 09/06/2024]
Abstract
Oncogenic mutations are abundant in the tissues of healthy individuals, but rarely form tumours1-3. Yet, the underlying protection mechanisms are largely unknown. To resolve these mechanisms in mouse mammary tissue, we use lineage tracing to map the fate of wild-type and Brca1-/-;Trp53-/- cells, and find that both follow a similar pattern of loss and spread within ducts. Clonal analysis reveals that ducts consist of small repetitive units of self-renewing cells that give rise to short-lived descendants. This offers a first layer of protection as any descendants, including oncogenic mutant cells, are constantly lost, thereby limiting the spread of mutations to a single stem cell-descendant unit. Local tissue remodelling during consecutive oestrous cycles leads to the cooperative and stochastic loss and replacement of self-renewing cells. This process provides a second layer of protection, leading to the elimination of most mutant clones while enabling the minority that by chance survive to expand beyond the stem cell-descendant unit. This leads to fields of mutant cells spanning large parts of the epithelial network, predisposing it for transformation. Eventually, clone expansion becomes restrained by the geometry of the ducts, providing a third layer of protection. Together, these mechanisms act to eliminate most cells that acquire somatic mutations at the expense of driving the accelerated expansion of a minority of cells, which can colonize large areas, leading to field cancerization.
Collapse
Affiliation(s)
- Marta Ciwinska
- VIB-KULeuven Centre for Cancer Biology, Department of Oncology, Leuven, Belgium
| | - Hendrik A Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hristina R Hristova
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Rolf Harkes
- Bioimaging Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nathalia S M Langedijk
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Stefan J Hutten
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renée X Menezes
- Biostatistics Centre and Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Stefan Prekovic
- Centre for Molecular Medicine, UMC Utrecht, Utrecht, the Netherlands
| | - Benjamin D Simons
- Gurdon Institute, University of Cambridge, Cambridge, UK.
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK.
| | | | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Zhang T, Zhao C, Li Y, Wu J, Wang F, Yu J, Wang Z, Gao Y, Zhao L, Liu Y, Yan Y, Li X, Gao H, Hu Z, Cui B, Li K. FGD5 in basal cells induces CXCL14 secretion that initiates a feedback loop to promote murine mammary epithelial growth and differentiation. Dev Cell 2024; 59:2085-2100.e9. [PMID: 38821057 DOI: 10.1016/j.devcel.2024.05.007] [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: 08/25/2022] [Revised: 12/22/2023] [Accepted: 05/09/2024] [Indexed: 06/02/2024]
Abstract
The interactions of environmental compartments with epithelial cells are essential for mammary gland development and homeostasis. Currently, the direct crosstalk between the endothelial niche and mammary epithelial cells remains poorly understood. Here, we show that faciogenital dysplasia 5 (FGD5) is enriched in mammary basal cells (BCs) and mediates critical interactions between basal and endothelial cells (ECs) in the mammary gland. Conditional deletion of Fgd5 reduced, whereas conditional knockin of Fgd5 increased, the engraftment and expansion of BCs, regulating ductal morphogenesis in the mammary gland. Mechanistically, murine mammary BC-expressed FGD5 inhibited the transcriptional activity of activating transcription factor 3 (ATF3), leading to subsequent transcriptional activation and secretion of CXCL14. Furthermore, activation of CXCL14/CXCR4/ERK signaling in primary murine mammary stromal ECs enhanced the expression of HIF-1α-regulated hedgehog ligands, which initiated a positive feedback loop to promote the function of BCs. Collectively, these findings identify functionally important interactions between BCs and the endothelial niche that occur through the FGD5/CXCL14/hedgehog axis.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chenxi Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yunxuan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Feng Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jinmei Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhenhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yang Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Luyao Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yechao Yan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xia Li
- Marine College, Shandong University, Weihai 264200, China
| | - Huan Gao
- Marine College, Shandong University, Weihai 264200, China
| | - Zhuowei Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Ke Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
23
|
Muthuswamy SK, Brugge JS. Organoid Cultures for the Study of Mammary Biology and Breast Cancer: The Promise and Challenges. Cold Spring Harb Perspect Med 2024; 14:a041661. [PMID: 38110241 PMCID: PMC11216180 DOI: 10.1101/cshperspect.a041661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
During the last decade, biomedical research has experienced a resurgence in the use of three-dimensional culture models for studies of normal and cancer biology. This resurgence has been driven by the development of models in which primary cells are grown in tissue-mimicking media and extracellular matrices to create organoid or organotypic cultures that more faithfully replicate the complex architecture and physiology of normal tissues and tumors. In addition, patient-derived tumor organoids preserve the three-dimensional organization and characteristics of the patient tumors ex vivo, becoming excellent preclinical models to supplement studies of tumor xenografts transplanted into immunocompromised mice. In this perspective, we provide an overview of how organoids are being used to investigate normal mammary biology and as preclinical models of breast cancer and discuss improvements that would enhance their utility and relevance to the field.
Collapse
Affiliation(s)
- Senthil K Muthuswamy
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland 20894, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Ludwig Center at Harvard, Harvard Medical School Boston, Boston, Massachusetts 02115, USA
| |
Collapse
|
24
|
Ma R, Feng D, Chen J, Zhou J, Xia K, Kong X, Hu G, Lu P. Targeting Tumor Heterogeneity by Breaking a Stem Cell and Epithelial Niche Interaction Loop. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307452. [PMID: 38708713 PMCID: PMC11234407 DOI: 10.1002/advs.202307452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/20/2024] [Indexed: 05/07/2024]
Abstract
Tumor heterogeneity, the presence of multiple distinct subpopulations of cancer cells between patients or among the same tumors, poses a major challenge to current targeted therapies. The way these different subpopulations interact among themselves and the stromal niche environment, and how such interactions affect cancer stem cell behavior has remained largely unknown. Here, it is shown that an FGF-BMP7-INHBA signaling positive feedback loop integrates interactions among different cell populations, including mammary gland stem cells, luminal epithelial and stromal fibroblast niche components not only in organ regeneration but also, with certain modifications, in cancer progression. The reciprocal dependence of basal stem cells and luminal epithelium is based on basal-derived BMP7 and luminal-derived INHBA, which promote their respective expansion, and is regulated by stromal-epithelial FGF signaling. Targeting this interaction loop, for example, by reducing the function of one or more of its components, inhibits organ regeneration and breast cancer progression. The results have profound implications for overcoming drug resistance because of tumor heterogeneity in future targeted therapies.
Collapse
Affiliation(s)
- Rongze Ma
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Deyi Feng
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
| | - Jing Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| | - Jiecan Zhou
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Kun Xia
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
| | - Xiangyin Kong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Pengfei Lu
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
25
|
Kannan N, Deshpande AJ. Connie J. Eaves (1944-2024). Nat Rev Cancer 2024; 24:443. [PMID: 38698125 DOI: 10.1038/s41568-024-00696-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Affiliation(s)
- Nagarajan Kannan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Aniruddha J Deshpande
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
26
|
Bai H, Liu X, Lin M, Meng Y, Tang R, Guo Y, Li N, Clarke MF, Cai S. Progressive senescence programs induce intrinsic vulnerability to aging-related female breast cancer. Nat Commun 2024; 15:5154. [PMID: 38886378 PMCID: PMC11183265 DOI: 10.1038/s41467-024-49106-2] [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: 06/16/2023] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
Cancer incidence escalates exponentially with advancing age; however, the underlying mechanism remains unclear. In this study, we build a chronological molecular clock at single-cell transcription level with a mammary stem cell-enriched population to depict physiological aging dynamics in female mice. We find that the mammary aging process is asynchronous and progressive, initiated by an early senescence program, succeeded by an entropic late senescence program with elevated cancer associated pathways, vulnerable to cancer predisposition. The transition towards senescence program is governed by a stem cell factor Bcl11b, loss of which accelerates mammary ageing with enhanced DMBA-induced tumor formation. We have identified a drug TPCA-1 that can rejuvenate mammary cells and significantly reduce aging-related cancer incidence. Our findings establish a molecular portrait of progressive mammary cell aging and elucidate the transcriptional regulatory network bridging mammary aging and cancer predisposition, which has potential implications for the management of cancer prevalence in the aged.
Collapse
Affiliation(s)
- Huiru Bai
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Xiaoqin Liu
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Meizhen Lin
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Yuan Meng
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Ruolan Tang
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Yajing Guo
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China
| | - Nan Li
- Westlake University High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Michael F Clarke
- Institute of Stem Cell and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Shang Cai
- Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
Khalil AA, Smits D, Haughton PD, Koorman T, Jansen KA, Verhagen MP, van der Net M, van Zwieten K, Enserink L, Jansen L, El-Gammal AG, Visser D, Pasolli M, Tak M, Westland D, van Diest PJ, Moelans CB, Roukens MG, Tavares S, Fortier AM, Park M, Fodde R, Gloerich M, Zwartkruis FJT, Derksen PW, de Rooij J. A YAP-centered mechanotransduction loop drives collective breast cancer cell invasion. Nat Commun 2024; 15:4866. [PMID: 38849373 PMCID: PMC11161601 DOI: 10.1038/s41467-024-49230-z] [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: 02/16/2022] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Dense and aligned Collagen I fibers are associated with collective cancer invasion led by protrusive tumor cells, leader cells. In some breast tumors, a population of cancer cells (basal-like cells) maintain several epithelial characteristics and express the myoepithelial/basal cell marker Keratin 14 (K14). Emergence of leader cells and K14 expression are regarded as interconnected events triggered by Collagen I, however the underlying mechanisms remain unknown. Using breast carcinoma organoids, we show that Collagen I drives a force-dependent loop, specifically in basal-like cancer cells. The feed-forward loop is centered around the mechanotransducer Yap and independent of K14 expression. Yap promotes a transcriptional program that enhances Collagen I alignment and tension, which further activates Yap. Active Yap is detected in invading breast cancer cells in patients and required for collective invasion in 3D Collagen I and in the mammary fat pad of mice. Our work uncovers an essential function for Yap in leader cell selection during collective cancer invasion.
Collapse
Affiliation(s)
- Antoine A Khalil
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Daan Smits
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter D Haughton
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Thijs Koorman
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karin A Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mathijs P Verhagen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Mirjam van der Net
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kitty van Zwieten
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lotte Enserink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lisa Jansen
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Abdelrahman G El-Gammal
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daan Visser
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Milena Pasolli
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Max Tak
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Denise Westland
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Guy Roukens
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sandra Tavares
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Anne-Marie Fortier
- Goodman Cancer Institute McGill University, Depts Biochemistry and Oncology, McGill University, Goodman Cancer Institute, Montréal, Canada
| | - Morag Park
- Goodman Cancer Institute McGill University, Depts Biochemistry and Oncology, McGill University, Goodman Cancer Institute, Montréal, Canada
| | - Riccardo Fodde
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martijn Gloerich
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fried J T Zwartkruis
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Patrick Wb Derksen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Johan de Rooij
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
28
|
Faraldo MM, Romagnoli M, Wallon L, Dubus P, Deugnier MA, Fre S. Alpha-6 integrin deletion delays the formation of Brca1/p53-deficient basal-like breast tumors by restricting luminal progenitor cell expansion. Breast Cancer Res 2024; 26:91. [PMID: 38835038 PMCID: PMC11151721 DOI: 10.1186/s13058-024-01851-4] [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: 12/19/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND The aberrant amplification of mammary luminal progenitors is at the origin of basal-like breast cancers associated with BRCA1 mutations. Integrins mediate cell-matrix adhesion and transmit mechanical and chemical signals that drive epithelial stem cell functions and regulate tumor progression, metastatic reactivation, and resistance to targeted therapies. Consistently, we have recently shown that laminin-binding integrins are essential for the expansion and differentiation of mammary luminal progenitors in physiological conditions. As over-expression of the laminin-binding α6 integrin (Itgα6) is associated with poor prognosis and reduced survival in breast cancer, we here investigate the role of Itgα6 in mammary tumorigenesis. METHODS We used Blg-Cre; Brca1F/F; Trp53F/F mice, a model that phenocopies human basal-like breast cancer with BRCA1 mutations. We generated mutant mice proficient or deficient in Itgα6 expression and followed tumor formation. Mammary tumors and pretumoral tissues were characterized by immunohistochemistry, flow cytometry, RT-qPCR, Western blotting and organoid cultures. Clonogenicity of luminal progenitors from preneoplastic glands was studied in 3D Matrigel cultures. RESULTS We show that Itga6 deletion favors activation of p16 cell cycle inhibitor in the preneoplastic tissue. Subsequently, the amplification of luminal progenitors, the cell of origin of Brca1-deficient tumors, is restrained in Itgα6-deficient gland. In addition, the partial EMT program operating in Brca1/p53-deficient epithelium is attenuated in the absence of Itgα6. As a consequence of these events, mammary tumor formation is delayed in Itgα6-deficient mice. After tumor formation, the lack of Itgα6 does not affect tumor growth but rather alters their differentiation, resulting in reduced expression of basal cell markers. CONCLUSIONS Our data indicate that Itgα6 has a pro-tumorigenic role in Blg-Cre; Brca1F/F; Trp53F/F mice developing basal-like mammary tumors. In particular, we reveal that Itgα6 is required for the luminal progenitor expansion and the aberrant partial EMT program that precedes the formation of BRCA1 deficient tumors.
Collapse
Affiliation(s)
- Marisa M Faraldo
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France.
| | - Mathilde Romagnoli
- Laboratory of Cell Biology and Cancer, CNRS UMR144, Institut Curie, PSL Research University, 75248, Paris, France
- Institut de Recherches Internationales Servier, 91190, Gif Sur Yvette, France
| | - Loane Wallon
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France
- Alacris Theranostics GmbH, 12489, Berlin, Germany
| | - Pierre Dubus
- Department of Histology and Pathology, Centre Hospitalier Universitaire de Bordeaux, 33000, Bordeaux, France
- BRIC U1312, INSERM, Bordeaux Institute of Oncology, Université de Bordeaux, 33000, Bordeaux, France
| | - Marie-Ange Deugnier
- Laboratory of Cell Biology and Cancer, CNRS UMR144, Institut Curie, PSL Research University, 75248, Paris, France
| | - Silvia Fre
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France.
| |
Collapse
|
29
|
Audet J, Berger M, Cashman J, Chalandon Y, Coulombel L, Flamant S, Ghaffari S, Lefort S, Lemoine F, Maguer-Satta V, Nicolini FE, Schmitt C, Sloma I, Turhan A. Pr Connie J. Eaves (1944–2024). Bull Cancer 2024:S0007-4551(24)00160-7. [PMID: 38760290 DOI: 10.1016/j.bulcan.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Affiliation(s)
- Julie Audet
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Marc Berger
- Hématologie biologique and EA 7453 CHELTER, CHU d'Estaing, Clermont-Ferrand, France; Université de Clermont Auvergne, Clermont-Ferrand, France
| | | | - Yves Chalandon
- Service d'hématologie, département d'oncologie, hôpitaux universitaires Genève (HUG), Genève, Suisse; Faculté de médecine, université de Genève, Genève, Suisse
| | | | - Stéphane Flamant
- Radiobiology and Regenerative Medicine Research Service, Radiobiology of Accidental Exposure Laboratory, IRSN PSE-SANTE/SERAMED/LRAcc, 31, avenue de la Division-Leclerc, 92260 Fontenay-aux-Roses, France
| | - Saghi Ghaffari
- Department of Cell, Developmental and Regenerative Biology and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, États-Unis; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, États-Unis
| | - Sylvain Lefort
- CNRS UMR5286, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; INSERM U1052, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; Department of Cancer Initiation and Tumor cell Identity, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; CRCL, universite Claude-Bernard Lyon 1, Lyon, France
| | - François Lemoine
- Sorbonne University Paris, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; INSERM U1052, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; Department of Cancer Initiation and Tumor cell Identity, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; CRCL, universite Claude-Bernard Lyon 1, Lyon, France; Centre Léon-Bérard, Lyon, France
| | - Franck E Nicolini
- INSERM U1052, centre de recherche en cancérologie de Lyon, centre Léon Bérard, Lyon, France; Hématologie clinique, centre Léon-Bérard, 28, rue Laennec, 69373 Lyon cedex 08, France.
| | - Christian Schmitt
- INSERM U. 659, faculté de médecine de Créteil, 8, rue du Général-Sarrail, 94010 Créteil, France
| | - Ivan Sloma
- Hematology Department, AP-HP, UPEC University, Henri-Mondor Hospital, Créteil, France
| | - Ali Turhan
- INSERM U935, service d'hématologie, hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| |
Collapse
|
30
|
Williams MJ, Oliphant MU, Au V, Liu C, Baril C, O'Flanagan C, Lai D, Beatty S, Van Vliet M, Yiu JC, O'Connor L, Goh WL, Pollaci A, Weiner AC, Grewal D, McPherson A, Moore M, Prabhakar V, Agarwal S, Garber JE, Dillon D, Shah SP, Brugge J, Aparicio S. Luminal breast epithelial cells from wildtype and BRCA mutation carriers harbor copy number alterations commonly associated with breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591587. [PMID: 38746396 PMCID: PMC11092623 DOI: 10.1101/2024.05.01.591587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Cancer-associated mutations have been documented in normal tissues, but the prevalence and nature of somatic copy number alterations and their role in tumor initiation and evolution is not well understood. Here, using single cell DNA sequencing, we describe the landscape of CNAs in >42,000 breast epithelial cells from women with normal or high risk of developing breast cancer. Accumulation of individual cells with one or two of a specific subset of CNAs (e.g. 1q gain and 16q, 22q, 7q, and 10q loss) is detectable in almost all breast tissues and, in those from BRCA1 or BRCA2 mutations carriers, occurs prior to loss of heterozygosity (LOH) of the wildtype alleles. These CNAs, which are among the most common associated with ductal carcinoma in situ (DCIS) and malignant breast tumors, are enriched almost exclusively in luminal cells not basal myoepithelial cells. Allele-specific analysis of the enriched CNAs reveals that each allele was independently altered, demonstrating convergent evolution of these CNAs in an individual breast. Tissues from BRCA1 or BRCA2 mutation carriers contain a small percentage of cells with extreme aneuploidy, featuring loss of TP53 , LOH of BRCA1 or BRCA2 , and multiple breast cancer-associated CNAs in addition to one or more of the common CNAs in 1q, 10q or 16q. Notably, cells with intermediate levels of CNAs are not detected, arguing against a stepwise gradual accumulation of CNAs. Overall, our findings demonstrate that chromosomal alterations in normal breast epithelium partially mirror those of established cancer genomes and are chromosome- and cell lineage-specific.
Collapse
|
31
|
Waas M, Khoo A, Tharmapalan P, McCloskey CW, Govindarajan M, Zhang B, Khan S, Waterhouse PD, Khokha R, Kislinger T. Droplet-based proteomics reveals CD36 as a marker for progenitors in mammary basal epithelium. CELL REPORTS METHODS 2024; 4:100741. [PMID: 38569541 PMCID: PMC11045875 DOI: 10.1016/j.crmeth.2024.100741] [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: 12/11/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
Deep proteomic profiling of rare cell populations has been constrained by sample input requirements. Here, we present DROPPS (droplet-based one-pot preparation for proteomic samples), an accessible low-input platform that generates high-fidelity proteomic profiles of 100-2,500 cells. By applying DROPPS within the mammary epithelium, we elucidated the connection between mitochondrial activity and clonogenicity, identifying CD36 as a marker of progenitor capacity in the basal cell compartment. We anticipate that DROPPS will accelerate biology-driven proteomic research for a multitude of rare cell populations.
Collapse
Affiliation(s)
- Matthew Waas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Amanda Khoo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Pirashaanthy Tharmapalan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Curtis W McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Meinusha Govindarajan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Bowen Zhang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Shahbaz Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Paul D Waterhouse
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
32
|
Prokakis E, Bamahmoud H, Jansari S, Fritsche L, Dietz A, Boshnakovska A, Rehling P, Johnsen SA, Gallwas J, Wegwitz F. USP22 supports the aggressive behavior of basal-like breast cancer by stimulating cellular respiration. Cell Commun Signal 2024; 22:120. [PMID: 38347585 PMCID: PMC10863169 DOI: 10.1186/s12964-023-01441-5] [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: 06/28/2023] [Accepted: 12/12/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequent tumor entity in women worldwide with a high chance of therapeutic response in early- and non-metastatic disease stages. Among all BC subtypes, triple-negative BC (TNBC) is the most challenging cancer subtype lacking effective molecular targets due to the particular enrichment of cancer stem cells (CSCs), frequently leading to a chemoresistant phenotype and metastasis. The Ubiquitin Specific Peptidase 22 (USP22) is a deubiquitinase that has been frequently associated with a CSC-promoting function and intimately implicated in resistance to conventional therapies, tumor relapse, metastasis and overall poor survival in a broad range of cancer entities, including BC. To date, though, the role of USP22 in TNBC has been only superficially addressed. METHODS The current study utilized the MMTV-cre, Usp22fl/fl transgenic mouse model to study the involvement of USP22 in the stem cell-like properties of the growing mammary tissue. Additionally, we combined high-throughput transcriptomic analyses with publicly available patient transcriptomic data and utilized TNBC culture models to decipher the functional role of USP22 in the CSC characteristics of this disease. RESULTS Interestingly, we identified that USP22 promotes CSC properties and drug tolerance by supporting the oxidative phosphorylation program, known to be largely responsible for the poor response to conventional therapies in this particularly aggressive BC subtype. CONCLUSIONS This study suggests a novel tumor-supportive role of USP22 in sustaining cellular respiration to facilitate the drug-tolerant behavior of HER2+-BC and TNBC cells. Therefore, we posit USP22 as a promising therapeutic target to optimize standard therapies and combat the aggressiveness of these malignancies. Video Abstract.
Collapse
Affiliation(s)
- Evangelos Prokakis
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany.
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany.
| | - Husam Bamahmoud
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Shaishavi Jansari
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Lena Fritsche
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Dietz
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Steven A Johnsen
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- The Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Julia Gallwas
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany.
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
33
|
Abstract
In the 160 years since Rudolf Virchow first postulated that neoplasia arises by the same law that regulates embryonic development, scientists have come to recognize the striking overlap between the molecular and cellular programs used by cancers and embryos. Advances in cancer biology and molecular techniques have further highlighted the similarities between carcinogenesis and embryogenesis, where cellular growth, differentiation, motility, and intercellular cross talk are mediated by common drivers and regulatory networks. This review highlights the many connections linking cancer biology and developmental biology to provide a deeper understanding of how a tissue's developmental history may both enable and constrain cancer cell evolution.
Collapse
Affiliation(s)
- Ben Z Stanger
- Division of Gastroenterology, Department of Medicine, Abramson Family Cancer Research Institute, and Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Geoffrey M Wahl
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA;
| |
Collapse
|
34
|
Sahu S, Sahoo S, Sullivan T, O'Sullivan TN, Turan S, Albaugh ME, Burkett S, Tran B, Salomon DS, Kozlov SV, Koehler KR, Jolly MK, Sharan SK. Spatiotemporal modulation of growth factors directs the generation of multilineage mouse embryonic stem cell-derived mammary organoids. Dev Cell 2024; 59:175-186.e8. [PMID: 38159568 PMCID: PMC10872289 DOI: 10.1016/j.devcel.2023.12.003] [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: 12/01/2022] [Revised: 09/20/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Ectodermal appendages, such as the mammary gland (MG), are thought to have evolved from hair-associated apocrine glands to serve the function of milk secretion. Through the directed differentiation of mouse embryonic stem cells (mESCs), here, we report the generation of multilineage ESC-derived mammary organoids (MEMOs). We adapted the skin organoid model, inducing the dermal mesenchyme to transform into mammary-specific mesenchyme via the sequential activation of Bone Morphogenetic Protein 4 (BMP4) and Parathyroid Hormone-related Protein (PTHrP) and inhibition of hedgehog (HH) signaling. Using single-cell RNA sequencing, we identified gene expression profiles that demonstrate the presence of mammary-specific epithelial cells, fibroblasts, and adipocytes. MEMOs undergo ductal morphogenesis in Matrigel and can reconstitute the MG in vivo. Further, we demonstrate that the loss of function in placode regulators LEF1 and TBX3 in mESCs results in impaired skin and MEMO generation. In summary, our MEMO model is a robust tool for studying the development of ectodermal appendages, and it provides a foundation for regenerative medicine and disease modeling.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | - Teresa Sullivan
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - T Norene O'Sullivan
- Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA
| | - Sevilay Turan
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Mary E Albaugh
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Sandra Burkett
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Bao Tran
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - David S Salomon
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Serguei V Kozlov
- Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA; Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Karl R Koehler
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Otolaryngology, Department of Plastic & Oral Surgery, and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | - Shyam K Sharan
- Mouse Cancer Genetics Program (MCGP), Centre for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Centre for Advanced Preclinical Research (CAPR), National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
35
|
Liu C, Xu Y, Yang G, Tao Y, Chang J, Wang S, Cheung TH, Chen J, Zeng YA. Niche inflammatory signals control oscillating mammary regeneration and protect stem cells from cytotoxic stress. Cell Stem Cell 2024; 31:89-105.e6. [PMID: 38141612 DOI: 10.1016/j.stem.2023.11.012] [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: 04/16/2023] [Revised: 09/03/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023]
Abstract
Stem cells are known for their resilience and enhanced activity post-stress. The mammary gland undergoes frequent remodeling and is subjected to recurring stress during the estrus cycle, but it remains unclear how mammary stem cells (MaSCs) respond to the stress and contribute to regeneration. We discovered that cytotoxic stress-induced activation of CD11c+ ductal macrophages aids stem cell survival and prevents differentiation. These macrophages boost Procr+ MaSC activity through IL1β-IL1R1-NF-κB signaling during the estrus cycle in an oscillating manner. Deleting IL1R1 in MaSCs results in stem cell loss and skewed luminal differentiation. Moreover, under cytotoxic stress from the chemotherapy agent paclitaxel, ductal macrophages secrete higher IL1β levels, promoting MaSC survival and preventing differentiation. Inhibiting IL1R1 sensitizes MaSCs to paclitaxel. Our findings reveal a recurring inflammatory process that regulates regeneration, providing insights into stress-induced inflammation and its impact on stem cell survival, potentially affecting cancer therapy efficacy.
Collapse
Affiliation(s)
- Chunye Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yishu Xu
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Guowei Yang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yu Tao
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiali Chang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shihui Wang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research Center for Systems Biology and Human Health, the State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518057, Guangdong, China
| | - Jianfeng Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
36
|
Beumer J, Clevers H. Hallmarks of stemness in mammalian tissues. Cell Stem Cell 2024; 31:7-24. [PMID: 38181752 PMCID: PMC10769195 DOI: 10.1016/j.stem.2023.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024]
Abstract
All adult tissues experience wear and tear. Most tissues can compensate for cell loss through the activity of resident stem cells. Although the cellular maintenance strategies vary greatly between different adult (read: postnatal) tissues, the function of stem cells is best defined by their capacity to replace lost tissue through division. We discuss a set of six complementary hallmarks that are key enabling features of this basic function. These include longevity and self-renewal, multipotency, transplantability, plasticity, dependence on niche signals, and maintenance of genome integrity. We discuss these hallmarks in the context of some of the best-understood adult stem cell niches.
Collapse
Affiliation(s)
- Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| | - Hans Clevers
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| |
Collapse
|
37
|
Caruso M, Saberiseyedabad K, Mourao L, Scheele CLGJ. A Decision Tree to Guide Human and Mouse Mammary Organoid Model Selection. Methods Mol Biol 2024; 2764:77-105. [PMID: 38393590 DOI: 10.1007/978-1-0716-3674-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Over the past 50 years, researchers from the mammary gland field have launched a collection of distinctive 3D cell culture systems to study multiple aspects of mammary gland physiology and disease. As our knowledge about the mammary gland evolves, more sophisticated 3D cell culture systems are required to answer more and more complex questions. Nowadays, morphologically complex mammary organoids can be generated in distinct 3D settings, along with reproduction of multiple aspects of the gland microenvironment. Yet, each 3D culture protocol comes with its advantages and limitations, where some culture systems are best suited to study stemness potential, whereas others are tailored towards the study of mammary gland morphogenesis. Therefore, prior to starting a 3D mammary culture experiment, it is important to consider and select the ideal culture model to address the biological question of interest. The number and technical requirements of novel 3D cell culture methods vastly increased over the past decades, making it currently challenging and time consuming to identify the best experimental testing. In this chapter, we provide a summary of the most promising murine and human 3D organoid models that are currently used in mammary gland biology research. For each model, we will provide a brief description of the protocol and an overview of the expected morphological outcome, the advantages of the model, and the potential pitfalls, to guide the reader to the best model of choice for specific applications.
Collapse
Affiliation(s)
- Marika Caruso
- VIB-KU Leuven Center for Cancer Biology, Department of Oncology, Leuven, Belgium
| | | | - Larissa Mourao
- VIB-KU Leuven Center for Cancer Biology, Department of Oncology, Leuven, Belgium
| | | |
Collapse
|
38
|
Ivanova E, Hue-Beauvais C, Chaulot-Talmon A, Castille J, Laubier J, De Casanove C, Aubert-Frambourg A, Germon P, Jammes H, Le Provost F. DNA methylation and gene expression changes in mouse mammary tissue during successive lactations: part I - the impact of inflammation. Epigenetics 2023; 18:2215633. [PMID: 37302099 PMCID: PMC10732689 DOI: 10.1080/15592294.2023.2215633] [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: 12/06/2022] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Mastitis is among the main reasons women cease breastfeeding, which leads to them supplementing breast milk with artificial formula. In farm animals, mastitis results in significant economic losses and the premature culling of some animals. Nevertheless, researchers do not know enough about the effect of inflammation on the mammary gland. This article discusses the changes to DNA methylation in mouse mammary tissue caused by lipopolysaccharide-induced inflammation (4 h post-injection of lipopolysaccharide). We analysed the expression of some genes related to mammary gland function, epigenetic regulation, and the immune response. The analysis focused on three comparisons: inflammation during the first lactation, inflammation during second lactation with no history of inflammation, and inflammation during second lactation with previous inflammation. We identified differentially methylated cytosines (DMCs), differentially methylated regions (DMRs), and some differentially expressed genes (DEGs) for each comparison. The three comparisons shared some DEGs; however, few DMCs and only one DMR were shared. These observations suggest that inflammation is one of several factors affecting epigenetic regulation during successive lactations. Furthermore, the comparison between animals in second lactation with and without inflammation, with no inflammation history during first lactation showed a different pattern compared to the other conditions in this experiment. This indicates that inflammation history plays an important role in determining epigenetic changes. The data presented in this study suggest that lactation rank and previous inflammation history are equally important when explaining mammary tissue gene expression and DNA methylation changes.Abbreviations: RRBS, reduced representation bisulfite sequencing; RT-qPCR, real-time quantitative polymerase chain reaction; MEC, mammary epithelial cells; TSS, transcription start site; TTS, transcription termination site; UTR, untranslated region; SINE, short interspersed nuclear element; LINE, long interspersed nuclear element; CGI, CpG island; DEG, differentially expressed gene; DMC, differentially methylated cytosine; DMR, differentially methylated region; GO term, gene ontology term; MF, molecular function; BP, biological process.
Collapse
Affiliation(s)
- E. Ivanova
- Université Paris-Saclay, INRAE, AgroParistech, GABI, France
| | | | - A. Chaulot-Talmon
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-En-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - J. Castille
- Université Paris-Saclay, INRAE, AgroParistech, GABI, France
| | - J Laubier
- Université Paris-Saclay, INRAE, AgroParistech, GABI, France
| | - C De Casanove
- Université Paris-Saclay, INRAE, AgroParistech, GABI, France
| | - A. Aubert-Frambourg
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-En-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - P. Germon
- INRAE, Université de Tours, ISP, Nouzilly, France
| | - H. Jammes
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-En-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - F. Le Provost
- Université Paris-Saclay, INRAE, AgroParistech, GABI, France
| |
Collapse
|
39
|
Yuan L, Xie S, Bai H, Liu X, Cai P, Lu J, Wang C, Lin Z, Li S, Guo Y, Cai S. Reconstruction of dynamic mammary mini gland in vitro for normal physiology and oncogenesis. Nat Methods 2023; 20:2021-2033. [PMID: 37919421 DOI: 10.1038/s41592-023-02039-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/22/2023] [Indexed: 11/04/2023]
Abstract
Organoid culture has been extensively exploited for normal tissue reconstruction and disease modeling. However, it is still challenging to establish organoids that mimic in vivo-like architecture, size and function under homeostatic conditions. Here we describe the development of a long-term adult stem cell-derived mammary mini gland culture system that supports robust three-dimensional outgrowths recapitulating the morphology, scale, cellular context and transcriptional heterogeneity of the normal mammary gland. The self-organization ability of stem cells and the stability of the outgrowths were determined by a coordinated combination of extracellular matrix, environmental signals and dynamic physiological cycles. We show that these mini glands were hormone responsive and could recapitulate the entire postnatal mammary development including puberty, estrus cycle, lactation and involution. We also observed that these mini glands maintained the presence of mammary stem cells and could also recapitulate the fate transition from embryonic bipotency to postnatal unipotency in lineage tracing assays. In addition, upon induction of oncogene expression in the mini glands, we observed tumor initiation in vitro and in vivo in a mouse model. Together, this study provides an experimental system that can support a dynamic miniature mammary gland for the study of physiologically relevant, complex biological processes.
Collapse
Affiliation(s)
- Lei Yuan
- Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shaofang Xie
- Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Huiru Bai
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xiaoqin Liu
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Pei Cai
- Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Jing Lu
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Chunhui Wang
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Disease Modeling Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Zuobao Lin
- Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shuying Li
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Disease Modeling Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yajing Guo
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shang Cai
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Disease Modeling Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
40
|
Ivanova E, Hue-Beauvais C, Chaulot-Talmon A, Castille J, Laubier J, De Casanove C, Aubert-Frambourg A, Germon P, Jammes H, Le Provost F. DNA methylation and gene expression changes in mouse mammary tissue during successive lactations: part II - the impact of lactation rank. Epigenetics 2023; 18:2215620. [PMID: 37219968 PMCID: PMC10208124 DOI: 10.1080/15592294.2023.2215620] [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: 12/01/2022] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023] Open
Abstract
Mastitis is among the main reasons women cease breastfeeding. In farm animals, mastitis results in significant economic losses and the premature culling of some animals. Nevertheless, the effect of inflammation on the mammary gland is not completely understood. This article discusses the changes to DNA methylation in mouse mammary tissue caused by lipopolysaccharide-induced inflammation after in vivo intramammary challenges and the differences in DNA methylation between 1st and 2nd lactations. Lactation rank induces 981 differential methylations of cytosines (DMCs) in mammary tissue. Inflammation in 1st lactation compared to inflammation in 2nd lactation results in the identification of 964 DMCs. When comparing inflammation in 1st vs. 2nd lactations with previous inflammation history, 2590 DMCs were identified. Moreover, Fluidigm PCR data show changes in the expression of several genes related to mammary function, epigenetic regulation, and the immune response. We show that the epigenetic regulation of two successive physiological lactations is not the same in terms of DNA methylation and that the effect of lactation rank on DNA methylation is stronger than that of the onset of inflammation. The conditions presented here show that few DMCs are shared between comparisons, suggesting a specific epigenetic response depending on lactation rank, the presence of inflammation, and even whether the cells had previously suffered inflammation. In the long term, this information could lead to a better understanding of the epigenetic regulation of lactation in both physiological and pathological conditions.Abbreviations: RRBS, reduced representation bisulphite sequencing; RT-qPCR, real-time quantitative polymerase chain reaction; MEC, mammary epithelial cells; MaSC, mammary stem cell; TSS, transcription start site; TTS, transcription termination site; UTR, untranslated region; SINE, short interspersed nuclear element; LINE, long interspersed nuclear element; CGI, CpG island; DEG, differentially expressed gene; DMC, differentially methylated cytosine; DMR, differentially methylated region; GO term, gene ontology term; MF, molecular function; BP, biological process.
Collapse
Affiliation(s)
- E Ivanova
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - C Hue-Beauvais
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - A Chaulot-Talmon
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - J Castille
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - J Laubier
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - C De Casanove
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - A Aubert-Frambourg
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - P Germon
- INRAE, Université de Tours, Nouzilly, France
| | - H Jammes
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - F Le Provost
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
41
|
Wicker MN, Wagner KU. Cellular Plasticity in Mammary Gland Development and Breast Cancer. Cancers (Basel) 2023; 15:5605. [PMID: 38067308 PMCID: PMC10705338 DOI: 10.3390/cancers15235605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Cellular plasticity is a phenomenon where cells adopt different identities during development and tissue homeostasis as a response to physiological and pathological conditions. This review provides a general introduction to processes by which cells change their identity as well as the current definition of cellular plasticity in the field of mammary gland biology. Following a synopsis of the evolving model of the hierarchical development of mammary epithelial cell lineages, we discuss changes in cell identity during normal mammary gland development with particular emphasis on the effect of the gestation cycle on the emergence of new cellular states. Next, we summarize known mechanisms that promote the plasticity of epithelial lineages in the normal mammary gland and highlight the importance of the microenvironment and extracellular matrix. A discourse of cellular reprogramming during the early stages of mammary tumorigenesis that follows focuses on the origin of basal-like breast cancers from luminal progenitors and oncogenic signaling networks that orchestrate diverse developmental trajectories of transforming epithelial cells. In addition to the epithelial-to-mesenchymal transition, we highlight events of cellular reprogramming during breast cancer progression in the context of intrinsic molecular subtype switching and the genesis of the claudin-low breast cancer subtype, which represents the far end of the spectrum of epithelial cell plasticity. In the final section, we will discuss recent advances in the design of genetically engineered models to gain insight into the dynamic processes that promote cellular plasticity during mammary gland development and tumorigenesis in vivo.
Collapse
Affiliation(s)
| | - Kay-Uwe Wagner
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, 4100 John R, EL01TM, Detroit, MI 48201, USA
| |
Collapse
|
42
|
Milevskiy MJ, Coughlan HD, Kane SR, Johanson TM, Kordafshari S, Chan WF, Tsai M, Surgenor E, Wilcox S, Allan RS, Chen Y, Lindeman GJ, Smyth GK, Visvader JE. Three-dimensional genome architecture coordinates key regulators of lineage specification in mammary epithelial cells. CELL GENOMICS 2023; 3:100424. [PMID: 38020976 PMCID: PMC10667557 DOI: 10.1016/j.xgen.2023.100424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 12/01/2023]
Abstract
Although lineage-specific genes have been identified in the mammary gland, little is known about the contribution of the 3D genome organization to gene regulation in the epithelium. Here, we describe the chromatin landscape of the three major epithelial subsets through integration of long- and short-range chromatin interactions, accessibility, histone modifications, and gene expression. While basal genes display exquisite lineage specificity via distal enhancers, luminal-specific genes show widespread promoter priming in basal cells. Cell specificity in luminal progenitors is largely mediated through extensive chromatin interactions with super-enhancers in gene-body regions in addition to interactions with polycomb silencer elements. Moreover, lineage-specific transcription factors appear to be controlled through cell-specific chromatin interactivity. Finally, chromatin accessibility rather than interactivity emerged as a defining feature of the activation of quiescent basal stem cells. This work provides a comprehensive resource for understanding the role of higher-order chromatin interactions in cell-fate specification and differentiation in the adult mouse mammary gland.
Collapse
Affiliation(s)
- Michael J.G. Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Hannah D. Coughlan
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Serena R. Kane
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Timothy M. Johanson
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Somayeh Kordafshari
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Wing Fuk Chan
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Minhsuang Tsai
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elliot Surgenor
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Stephen Wilcox
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Rhys S. Allan
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Yunshun Chen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Geoffrey J. Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
- Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC 3050, Australia
| | - Gordon K. Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jane E. Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
43
|
Gray GK, Girnius N, Kuiken HJ, Henstridge AZ, Brugge JS. Single-cell and spatial analyses reveal a tradeoff between murine mammary proliferation and lineage programs associated with endocrine cues. Cell Rep 2023; 42:113293. [PMID: 37858468 PMCID: PMC10840493 DOI: 10.1016/j.celrep.2023.113293] [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: 05/25/2023] [Revised: 08/25/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023] Open
Abstract
Although distinct epithelial cell types have been distinguished in glandular tissues such as the mammary gland, the extent of heterogeneity within each cell type and the degree of endocrine control of this diversity across development are incompletely understood. By combining mass cytometry and cyclic immunofluorescence, we define a rich array of murine mammary epithelial cell subtypes associated with puberty, the estrous cycle, and sex. These subtypes are differentially proliferative and spatially segregate distinctly in adult versus pubescent glands. Further, we identify systematic suppression of lineage programs at the protein and RNA levels as a common feature of mammary epithelial expansion during puberty, the estrous cycle, and gestation and uncover a pervasive enrichment of ribosomal protein genes in luminal cells elicited specifically during progesterone-dominant expansionary periods. Collectively, these data expand our knowledge of murine mammary epithelial heterogeneity and connect endocrine-driven epithelial expansion with lineage suppression.
Collapse
Affiliation(s)
- G Kenneth Gray
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; The Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Hendrik J Kuiken
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Aylin Z Henstridge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Kim H, Aliar K, Tharmapalan P, McCloskey CW, Kuttanamkuzhi A, Grünwald BT, Palomero L, Mahendralingam MJ, Waas M, Mer AS, Elliott MJ, Zhang B, Al-Zahrani KN, Langille ER, Parsons M, Narala S, Hofer S, Waterhouse PD, Hakem R, Haibe-Kains B, Kislinger T, Schramek D, Cescon DW, Pujana MA, Berman HK, Khokha R. Differential DNA damage repair and PARP inhibitor vulnerability of the mammary epithelial lineages. Cell Rep 2023; 42:113256. [PMID: 37847590 DOI: 10.1016/j.celrep.2023.113256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 09/02/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
It is widely assumed that all normal somatic cells can equally perform homologous recombination (HR) and non-homologous end joining in the DNA damage response (DDR). Here, we show that the DDR in normal mammary gland inherently depends on the epithelial cell lineage identity. Bioinformatics, post-irradiation DNA damage repair kinetics, and clonogenic assays demonstrated luminal lineage exhibiting a more pronounced DDR and HR repair compared to the basal lineage. Consequently, basal progenitors were far more sensitive to poly(ADP-ribose) polymerase inhibitors (PARPis) in both mouse and human mammary epithelium. Furthermore, PARPi sensitivity of murine and human breast cancer cell lines as well as patient-derived xenografts correlated with their molecular resemblance to the mammary progenitor lineages. Thus, mammary epithelial cells are intrinsically divergent in their DNA damage repair capacity and PARPi vulnerability, potentially influencing the clinical utility of this targeted therapy.
Collapse
Affiliation(s)
- Hyeyeon Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Kazeera Aliar
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Pirashaanthy Tharmapalan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Curtis W McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Barbara T Grünwald
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Luis Palomero
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Catalonia, Spain
| | - Mathepan J Mahendralingam
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Matthew Waas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Arvind S Mer
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mitchell J Elliott
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Bowen Zhang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Khalid N Al-Zahrani
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Ellen R Langille
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael Parsons
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Swami Narala
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stefan Hofer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Paul D Waterhouse
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Miquel A Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Catalonia, Spain
| | - Hal K Berman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2N2, Canada.
| |
Collapse
|
45
|
Bouamar H, Broome LE, Lathrop KI, Jatoi I, Brenner AJ, Nazarullah A, Gorena KM, Garcia M, Chen Y, Kaklamani V, Sun LZ. mTOR inhibition abrogates human mammary stem cells and early breast cancer progression markers. Breast Cancer Res 2023; 25:131. [PMID: 37904250 PMCID: PMC10614399 DOI: 10.1186/s13058-023-01727-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/04/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Mammary physiology is distinguished in containing adult stem/progenitor cells that are actively amending the breast tissue throughout the reproductive lifespan of women. Despite their importance in both mammary gland development, physiological maintenance, and reproduction, the exact role of mammary stem/progenitor cells in mammary tumorigenesis has not been fully elucidated in humans or animal models. The implications of modulating adult stem/progenitor cells in women could lead to a better understanding of not only their function, but also toward possible breast cancer prevention led us to evaluate the efficacy of rapamycin in reducing mammary stem/progenitor cell activity and malignant progression markers. METHODS We analyzed a large number of human breast tissues for their basal and luminal cell composition with flow cytometry and their stem and progenitor cell function with sphere formation assay with respect to age and menopausal status in connection with a clinical study (NCT02642094) involving a low-dose (2 mg/day) and short-term (5-7 days) treatment of the mTOR inhibitor sirolimus. The expression of biomarkers in biopsies and surgical breast samples were measured with quantitative analysis of immunohistochemistry. RESULTS Sirolimus treatment significantly abrogated mammary stem cell activity, particularly in postmenopausal patients. It did not affect the frequency of luminal progenitors but decreased their self-renewal capacity. While sirolimus had no effect on basal cell population, it decreased luminal cell population, particularly in postmenopausal patients. It also significantly diminished prognostic biomarkers associated with breast cancer progression from ductal carcinoma in situ to invasive breast cancer including p16INK4A, COX-2, and Ki67, as well as markers of the senescence-associated secretary phenotype, thereby possibly functioning in preventing early breast cancer progression. CONCLUSION Overall, these findings indicate a link from mTOR signaling to mammary stem and progenitor cell activity and cancer progression. Trial registration This study involves a clinical trial registered under the ClinicalTrials.gov identifier NCT02642094 registered December 30, 2015.
Collapse
Affiliation(s)
- Hakim Bouamar
- Department of Cell Systems and Anatomy, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Larry Esteban Broome
- Department of Cell Systems and Anatomy, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kate Ida Lathrop
- Department of Medicine, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ismail Jatoi
- Department of Surgery, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Andrew Jacob Brenner
- Department of Medicine, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Alia Nazarullah
- Department of Pathology, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Karla Moncada Gorena
- Flow Cytometry Core Facility, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael Garcia
- Department of Medicine, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Department of Population Health Sciences, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Greheey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Virginia Kaklamani
- Department of Medicine, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
46
|
Rocha AS, Collado-Solé A, Graña-Castro O, Redondo-Pedraza J, Soria-Alcaide G, Cordero A, Santamaría PG, González-Suárez E. Luminal Rank loss decreases cell fitness leading to basal cell bipotency in parous mammary glands. Nat Commun 2023; 14:6213. [PMID: 37813842 PMCID: PMC10562464 DOI: 10.1038/s41467-023-41741-5] [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: 07/28/2022] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
Rank signaling pathway regulates mammary gland homeostasis and epithelial cell differentiation. Although Rank receptor is expressed by basal cells and luminal progenitors, its role in each individual cell lineage remains unclear. By combining temporal/lineage specific Rank genetic deletion with lineage tracing techniques, we found that loss of luminal Rank reduces the luminal progenitor pool and leads to aberrant alveolar-like differentiation with high protein translation capacity in virgin mammary glands. These Rank-deleted luminal cells are unable to expand during the first pregnancy, leading to lactation failure and impairment of protein synthesis potential in the parous stage. The unfit parous Rank-deleted luminal cells in the alveoli are progressively replaced by Rank-proficient cells early during the second pregnancy, thereby restoring lactation. Transcriptomic analysis and functional assays point to the awakening of basal bipotency after pregnancy by the induction of Rank/NF-κB signaling in basal parous cell to restore lactation and tissue homeostasis.
Collapse
Affiliation(s)
- Ana Sofia Rocha
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.
| | | | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA-Nemesio Díez), School of Medicine, San Pablo-CEU University, CEU Universities, Boadilla del Monte, Madrid, Spain
| | | | | | - Alex Cordero
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | | | - Eva González-Suárez
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.
- Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| |
Collapse
|
47
|
Fan Y, Jin L, He Z, Wei T, Luo T, Zhang J, Liu C, Dai C, A C, Liang Y, Tao X, Lv X, Gu Y, Li M. A cell transcriptomic profile provides insights into adipocytes of porcine mammary gland across development. J Anim Sci Biotechnol 2023; 14:126. [PMID: 37805503 PMCID: PMC10560433 DOI: 10.1186/s40104-023-00926-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/03/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Studying the composition and developmental mechanisms in mammary gland is crucial for healthy growth of newborns. The mammary gland is inherently heterogeneous, and its physiological function dependents on the gene expression of multiple cell types. Most studies focused on epithelial cells, disregarding the role of neighboring adipocytes. RESULTS Here, we constructed the largest transcriptomic dataset of porcine mammary gland cells thus far. The dataset captured 126,829 high-quality nuclei from physiological mammary glands across five developmental stages (d 90 of gestation, G90; d 0 after lactation, L0; d 20 after lactation, L20; 2 d post natural involution, PI2; 7 d post natural involution, PI7). Seven cell types were identified, including epithelial cells, adipocytes, endothelial cells, fibroblasts cells, immune cells, myoepithelial cells and precursor cells. Our data indicate that mammary glands at different developmental stages have distinct phenotypic and transcriptional signatures. During late gestation (G90), the differentiation and proliferation of adipocytes were inhibited. Meanwhile, partly epithelial cells were completely differentiated. Pseudo-time analysis showed that epithelial cells undergo three stages to achieve lactation, including cellular differentiation, hormone sensing, and metabolic activation. During lactation (L0 and L20), adipocytes area accounts for less than 0.5% of mammary glands. To maintain their own survival, the adipocyte exhibited a poorly differentiated state and a proliferative capacity. Epithelial cells initiate lactation upon hormonal stimulation. After fulfilling lactation mission, their undergo physiological death under high intensity lactation. Interestingly, the physiological dead cells seem to be actively cleared by immune cells via CCL21-ACKR4 pathway. This biological process may be an important mechanism for maintaining homeostasis of the mammary gland. During natural involution (PI2 and PI7), epithelial cell populations dedifferentiate into mesenchymal stem cells to maintain the lactation potential of mammary glands for the next lactation cycle. CONCLUSION The molecular mechanisms of dedifferentiation, proliferation and redifferentiation of adipocytes and epithelial cells were revealed from late pregnancy to natural involution. This cell transcriptomic profile constitutes an essential reference for future studies in the development and remodeling of the mammary gland at different stages.
Collapse
Affiliation(s)
- Yongliang Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu, 610041 China
| | - Long Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Zhiping He
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610000 China
| | - Tiantian Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Tingting Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Jiaman Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Can Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Changjiu Dai
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Chao A
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Yan Liang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610000 China
| | - Xuan Tao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610000 China
| | - Xuebin Lv
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610000 China
| | - Yiren Gu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu, 610041 China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610000 China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| |
Collapse
|
48
|
Pérez-González A, Bévant K, Blanpain C. Cancer cell plasticity during tumor progression, metastasis and response to therapy. NATURE CANCER 2023; 4:1063-1082. [PMID: 37537300 PMCID: PMC7615147 DOI: 10.1038/s43018-023-00595-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 06/01/2023] [Indexed: 08/05/2023]
Abstract
Cell plasticity represents the ability of cells to be reprogrammed and to change their fate and identity, enabling homeostasis restoration and tissue regeneration following damage. Cell plasticity also contributes to pathological conditions, such as cancer, enabling cells to acquire new phenotypic and functional features by transiting across distinct cell states that contribute to tumor initiation, progression, metastasis and resistance to therapy. Here, we review the intrinsic and extrinsic mechanisms driving cell plasticity that promote tumor growth and proliferation as well as metastasis and drug tolerance. Finally, we discuss how cell plasticity could be exploited for anti-cancer therapy.
Collapse
Affiliation(s)
- Andrea Pérez-González
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Kevin Bévant
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- WELBIO, ULB, Bruxelles, Belgium.
| |
Collapse
|
49
|
van Amerongen R, Bentires-Alj M, van Boxtel AL, Clarke RB, Fre S, Suarez EG, Iggo R, Jechlinger M, Jonkers J, Mikkola ML, Koledova ZS, Sørlie T, Vivanco MDM. Imagine beyond: recent breakthroughs and next challenges in mammary gland biology and breast cancer research. J Mammary Gland Biol Neoplasia 2023; 28:17. [PMID: 37450065 PMCID: PMC10349020 DOI: 10.1007/s10911-023-09544-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/25/2023] [Indexed: 07/18/2023] Open
Abstract
On 8 December 2022 the organizing committee of the European Network for Breast Development and Cancer labs (ENBDC) held its fifth annual Think Tank meeting in Amsterdam, the Netherlands. Here, we embraced the opportunity to look back to identify the most prominent breakthroughs of the past ten years and to reflect on the main challenges that lie ahead for our field in the years to come. The outcomes of these discussions are presented in this position paper, in the hope that it will serve as a summary of the current state of affairs in mammary gland biology and breast cancer research for early career researchers and other newcomers in the field, and as inspiration for scientists and clinicians to move the field forward.
Collapse
Affiliation(s)
- Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| | - Mohamed Bentires-Alj
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Robert B Clarke
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Silvia Fre
- Institut Curie, Genetics and Developmental Biology Department, PSL Research University, CNRS UMR3215, U93475248, InsermParis, France
| | - Eva Gonzalez Suarez
- Transformation and Metastasis Laboratory, Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Richard Iggo
- INSERM U1312, University of Bordeaux, 33076, Bordeaux, France
| | - Martin Jechlinger
- Cell Biology and Biophysics Department, EMBL, Heidelberg, Germany
- Molit Institute of Personalized Medicine, Heilbronn, Germany
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marja L Mikkola
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, P.O.B. 56, 00014, Helsinki, Finland
| | - Zuzana Sumbalova Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Maria dM Vivanco
- Cancer Heterogeneity Lab, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Technological Park Bizkaia, 48160, Derio, Spain
| |
Collapse
|
50
|
Tenreiro MF, Branco MA, Cotovio JP, Cabral JMS, Fernandes TG, Diogo MM. Advancing organoid design through co-emergence, assembly, and bioengineering. Trends Biotechnol 2023; 41:923-938. [PMID: 36653200 DOI: 10.1016/j.tibtech.2022.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023]
Abstract
Human adult stem cells and patient-derived induced pluripotent stem cells represent promising tools to understand human biology, development, and disease. Under a permissive environment, stem cell derivatives can self-organize and reconstruct their native milieu, resulting in the creation of organ-like entities known as organoids. Although organoids represent a breakthrough in the stem cell field, there are still considerable shortcomings preventing their widespread use, namely their variability, limited function, and reductionist size. In the past few years, sophisticated methodologies have been proposed to allow the design of organoids with improved biological fidelity and physiological relevance. Here, we summarize these emerging technologies and provide insights into how they can be utilized to fulfill the potential of stem cells.
Collapse
Affiliation(s)
- Miguel F Tenreiro
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Mariana A Branco
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - João P Cotovio
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.
| |
Collapse
|