1
|
Liu S, Cao H, Wang Z, Zhu J, An X, Zhang L, Song Y. Single-cell transcriptomics reveals extracellular matrix remodeling and collagen dynamics during lactation in sheep mammary gland. Int J Biol Macromol 2025; 312:143669. [PMID: 40319976 DOI: 10.1016/j.ijbiomac.2025.143669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland is a dynamic organ with diverse cell populations that maintain glandular homeostasis, particularly during lactation. However, the cellular architecture and molecular mechanisms underlying lactational remodeling in the sheep mammary gland remain incompletely understood. Given similarities in mammary stromal structure, sheep serve as a valuable model for studying lactational changes relevant to the human breast, which experiences collagen loss and sagging during lactation. Utilizing single-cell transcriptomics (scRNA-seq), we mapped the sheep mammary gland's cellular landscape at postpartum days 60 and 150, identifying seven major cell types, including six distinct epithelial clusters. These clusters revealed differentiation among luminal progenitors, hormone-sensing, and myoepithelial cells across peak and late lactation stages. Transcriptomic analysis highlighted pivotal roles for epithelial integrity and ECM remodeling, with myoepithelial cells centrally involved in these processes. We observed significant collagen remodeling driven by fibroblast-epithelial crosstalk and ECM reorganization during late lactation. Comparative analysis with human mammary epithelial cells showed conserved basal and myoepithelial cell populations, while luminal cells diverged across species. This study provides insights into lactation biology and ECM remodeling, offering a framework to inform future studies on lactational adaptation and its implications for human health.
Collapse
Affiliation(s)
- Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhanhang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
2
|
Downie JM, Musich RJ, Geraghty CM, Caraballo A, He S, Khawaled S, Lachut K, Zhou JY, Yilmaz ÖH, Stappenbeck TS, Chan AT, Drew DA. Droplet vs Picowell: Considerations for Single-cell Transcriptomic Profiling of Human Colon Biopsies. Cell Mol Gastroenterol Hepatol 2025:101503. [PMID: 40222388 DOI: 10.1016/j.jcmgh.2025.101503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Affiliation(s)
- Jonathan M Downie
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ryan J Musich
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
| | - Connor M Geraghty
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Alexander Caraballo
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Shijie He
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Saleh Khawaled
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Kylor Lachut
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Julie Y Zhou
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - David A Drew
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
3
|
Downie JM, Musich RJ, Geraghty CM, Caraballo A, He S, Khawaled S, Lachut K, Long T, Zhou JY, Yilmaz OH, Stappenbeck T, Chan AT, Drew DA. Optimizing single-cell RNA sequencing methods for human colon biopsies: droplet-based vs. picowell-based platforms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600526. [PMID: 38979379 PMCID: PMC11230261 DOI: 10.1101/2024.06.24.600526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background & Aims Single-cell RNA sequencing (scRNA) has empowered many insights into gastrointestinal microenvironments. However, profiling human biopsies using droplet-based scRNA (D-scRNA) is challenging since it requires immediate processing to minimize epithelial cell damage. In contrast, picowell-based (P-scRNA) platforms permit short-term frozen storage before sequencing. We compared P- and D-scRNA platforms on cells derived from human colon biopsies. Methods Endoscopic rectosigmoid mucosal biopsies were obtained from two adults and conducted D-scRNA (10X Chromium) and P-scRNA (Honeycomb HIVE) in parallel using an individual's pool of single cells (> 10,000 cells/participant). Three experiments were performed to evaluate 1) P-scRNA with cells under specific storage conditions (immediately processed [fresh], vs. frozen at -20C vs. -80C [2 weeks]); 2) fresh P-scRNA versus fresh D-scRNA; and 3) P-scRNA stored at -80C with fresh D-scRNA. Results Significant recovery of loaded cells was achieved for fresh (80.9%) and -80C (48.5%) P-scRNA and D-scRNA (76.6%), but not -20C P-scRNA (3.7%). However, D-scRNA captures more typeable cells among recovered cells (71.5% vs. 15.8% Fresh and 18.4% -80C P-scRNA), and these cells exhibit higher gene coverage at the expense of higher mitochondrial read fractions across most cell types. Cells profiled using D-scRNA demonstrated more consistent gene expression profiles among the same cell type than those profiled using P-scRNA. Significant intra-cell-type differences were observed in profiled gene classes across platforms. Conclusions Our results highlight non-overlapping advantages of P-scRNA and D-scRNA and underscore the need for innovation to enable high-fidelity capture of colonic epithelial cells. The platform-specific variation highlights the challenges of maintaining rigor and reproducibility across studies that use different platforms.
Collapse
|
4
|
Feng X, Tong L, Ma L, Mu T, Yu B, Ma R, Li J, Wang C, Zhang J, Gu Y. Mining key circRNA-associated-ceRNA networks for milk fat metabolism in cows with varying milk fat percentages. BMC Genomics 2024; 25:323. [PMID: 38561663 PMCID: PMC10983688 DOI: 10.1186/s12864-024-10252-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Cow milk fat is an essential indicator for evaluating and measuring milk quality and cow performance. Growing research has identified the molecular functions of circular RNAs (circRNAs) necessary for mammary gland development and lactation in mammals. METHOD The present study analyzed circRNA expression profiling data in mammary epithelial cells (MECs) from cows with highly variable milk fat percentage (MFP) using differential expression analysis and weighted gene co-expression network analysis (WGCNA). RESULTS A total of 309 differentially expressed circRNAs (DE-circRNAs) were identified in the high and low MFP groups. WGCNA analysis revealed that the pink module was significantly associated with MFP (r = - 0.85, P = 0.007). Parental genes of circRNAs in this module were enriched mainly in lipid metabolism-related signaling pathways, such as focal adhesion, ECM-receptor interaction, adherens junction and AMPK. Finally, six DE-circRNAs were screened from the pink module: circ_0010571, circ_0007797, circ_0002746, circ_0003052, circ_0004319, and circ_0012840. Among them, circ_0002746, circ_0003052, circ_0004319, and circ_0012840 had circular structures and were highly expressed in mammary tissues. Subcellular localization revealed that these four DE-circRNAs may play a regulatory role in the mammary glands of dairy cows, mainly as competitive endogenous RNAs (ceRNAs). Seven hub target genes (GNB1, GNG2, PLCB1, PLCG1, ATP6V0C, NDUFS4, and PIGH) were obtained by constructing the regulatory network of their ceRNAs and then analyzed by CytoHubba and MCODE plugins in Cytoscape. Functional enrichment analysis revealed that these genes are crucial and most probable ceRNA regulators in milk fat metabolism. CONCLUSIONS Our study identified several vital circRNAs and ceRNAs affecting milk fat synthesis, providing new research ideas and a theoretical basis for cow lactation, milk quality, and breed improvement.
Collapse
Affiliation(s)
- Xiaofang Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Lijia Tong
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Lina Ma
- NingXia Academy of Agriculture and Forestry Sciences, 750002, Yinchuan, China
| | - Tong Mu
- School of Life Science, Yan'an University, 716000, Yanan, China
| | - Baojun Yu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Ruoshuang Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Jiwei Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Chuanchuan Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Juan Zhang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China.
| | - Yaling Gu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| |
Collapse
|
5
|
Suresh RV, Deng B, Gebremicale Y, Roche K, Miura K, Long C. Mesenchymal stem cells of the bone marrow raise infectivity of Plasmodium falciparum gametocytes. mBio 2023; 14:e0223223. [PMID: 37909740 PMCID: PMC10746266 DOI: 10.1128/mbio.02232-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE While prior research has established that Plasmodium gametocytes sequester in the bone marrow and can influence resident stem cells, the question of why they would choose this compartment and these cells remained a mystery. This study, for the first time, shows that being in the presence of mesenchymal stem cells (MSCs) alters the biology of the P. falciparum parasite and makes it more infectious to mosquitoes, hinting at novel mechanisms in its life cycle. This method also facilitates mosquito infections with field isolated parasites, affording research teams new infection models with parasites, which are challenging to infect into mosquitos using conventional culture methods. Finally, our findings that MSC-conditioned medium can also raise infectivity open avenues of investigation into mechanisms involved but can also serve as a practical tool for researchers hoping to increase oocyst yields.
Collapse
Affiliation(s)
- Ragavan Varadharajan Suresh
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Bingbing Deng
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Yonas Gebremicale
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Kyle Roche
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Carole Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
6
|
Kumar T, Nee K, Wei R, He S, Nguyen QH, Bai S, Blake K, Pein M, Gong Y, Sei E, Hu M, Casasent AK, Thennavan A, Li J, Tran T, Chen K, Nilges B, Kashikar N, Braubach O, Ben Cheikh B, Nikulina N, Chen H, Teshome M, Menegaz B, Javaid H, Nagi C, Montalvan J, Lev T, Mallya S, Tifrea DF, Edwards R, Lin E, Parajuli R, Hanson S, Winocour S, Thompson A, Lim B, Lawson DA, Kessenbrock K, Navin N. A spatially resolved single-cell genomic atlas of the adult human breast. Nature 2023; 620:181-191. [PMID: 37380767 PMCID: PMC11443819 DOI: 10.1038/s41586-023-06252-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 05/23/2023] [Indexed: 06/30/2023]
Abstract
The adult human breast is comprised of an intricate network of epithelial ducts and lobules that are embedded in connective and adipose tissue1-3. Although most previous studies have focused on the breast epithelial system4-6, many of the non-epithelial cell types remain understudied. Here we constructed the comprehensive Human Breast Cell Atlas (HBCA) at single-cell and spatial resolution. Our single-cell transcriptomics study profiled 714,331 cells from 126 women, and 117,346 nuclei from 20 women, identifying 12 major cell types and 58 biological cell states. These data reveal abundant perivascular, endothelial and immune cell populations, and highly diverse luminal epithelial cell states. Spatial mapping using four different technologies revealed an unexpectedly rich ecosystem of tissue-resident immune cells, as well as distinct molecular differences between ductal and lobular regions. Collectively, these data provide a reference of the adult normal breast tissue for studying mammary biology and diseases such as breast cancer.
Collapse
Affiliation(s)
- Tapsi Kumar
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin Nee
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Runmin Wei
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Siyuan He
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quy H Nguyen
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Shanshan Bai
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Kerrigan Blake
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
- Math, Computational & Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Maren Pein
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Yanwen Gong
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- Math, Computational & Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Emi Sei
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Min Hu
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Anna K Casasent
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Aatish Thennavan
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Jianzhuo Li
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Tuan Tran
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | - Hui Chen
- Department of Pathology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Mediget Teshome
- Department of Breast Surgical Oncology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Menegaz
- Department of Pathology and Immunology, Baylor Medical College, Houston, TX, USA
| | - Huma Javaid
- Department of Pathology and Immunology, Baylor Medical College, Houston, TX, USA
| | - Chandandeep Nagi
- Department of Pathology and Immunology, Baylor Medical College, Houston, TX, USA
| | - Jessica Montalvan
- Department of Pathology and Immunology, Baylor Medical College, Houston, TX, USA
| | - Tatyana Lev
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
- Math, Computational & Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Sharmila Mallya
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Delia F Tifrea
- Chao Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| | - Robert Edwards
- Chao Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| | - Erin Lin
- Chao Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| | - Ritesh Parajuli
- Chao Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| | - Summer Hanson
- Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | | | | | - Bora Lim
- Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Devon A Lawson
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| | - Nicholas Navin
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Kumar T, Nee K, Wei R, He S, Nguyen QH, Bai S, Blake K, Gong Y, Pein M, Sei E, Hu M, Casasent A, Thennavan A, Li J, Tran T, Chen K, Nilges B, Kashikar N, Braubach O, Cheikh BB, Nikulina N, Chen H, Teshome M, Menegaz B, Javaid H, Nagi C, Montalvan J, Tifrea DF, Edwards R, Lin E, Parajuli R, Winocour S, Thompson A, Lim B, Lawson DA, Kessenbrock K, Navin N. A spatially resolved single cell genomic atlas of the adult human breast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.22.537946. [PMID: 37163043 PMCID: PMC10168262 DOI: 10.1101/2023.04.22.537946] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The adult human breast comprises an intricate network of epithelial ducts and lobules that are embedded in connective and adipose tissue. While previous studies have mainly focused on the breast epithelial system, many of the non-epithelial cell types remain understudied. Here, we constructed a comprehensive Human Breast Cell Atlas (HBCA) at single-cell and spatial resolution. Our single-cell transcriptomics data profiled 535,941 cells from 62 women, and 120,024 nuclei from 20 women, identifying 11 major cell types and 53 cell states. These data revealed abundant pericyte, endothelial and immune cell populations, and highly diverse luminal epithelial cell states. Our spatial mapping using three technologies revealed an unexpectedly rich ecosystem of tissue-resident immune cells in the ducts and lobules, as well as distinct molecular differences between ductal and lobular regions. Collectively, these data provide an unprecedented reference of adult normal breast tissue for studying mammary biology and disease states such as breast cancer.
Collapse
|
8
|
Mironov A, Fisher M, Narayanan P, Elsayed R, Karabulutoglu M, Akhtar N. Rac1 controls cell turnover and reversibility of the involution process in postpartum mammary glands. PLoS Biol 2023; 21:e3001583. [PMID: 36656812 PMCID: PMC9851507 DOI: 10.1371/journal.pbio.3001583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 12/11/2022] [Indexed: 01/20/2023] Open
Abstract
Cell turnover in adult tissues is essential for maintaining tissue homeostasis over a life span and for inducing the morphological changes associated with the reproductive cycle. However, the underlying mechanisms that coordinate the balance of cell death and proliferation remain unsolved. Using the mammary gland, we have discovered that Rac1 acts as a nexus to control cell turnover. Postlactational tissue regression is characterised by the death of milk secreting alveoli, but the process is reversible within the first 48 h if feeding recommences. In mice lacking epithelial Rac1, alveolar regression was delayed. This defect did not result from failed cell death but rather increased cell turnover. Fitter progenitor cells inappropriately divided, regenerating the alveoli, but cell death also concomitantly accelerated. We discovered that progenitor cell hyperproliferation was linked to nonautonomous effects of Rac1 deletion on the macrophageal niche with heightened inflammation. Moreover, loss of Rac1 impaired cell death with autophagy but switched the cell death route to apoptosis. Finally, mammary gland reversibility failed in the absence of Rac1 as the alveoli failed to recommence lactation upon resuckling.
Collapse
Affiliation(s)
- Aleksandr Mironov
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Matthew Fisher
- The Bateson Centre and Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Priya Narayanan
- The Bateson Centre and Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Randa Elsayed
- The Bateson Centre and Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Melis Karabulutoglu
- The Bateson Centre and Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Nasreen Akhtar
- The Bateson Centre and Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Feng X, Cai Z, Gu Y, Mu T, Yu B, Ma R, Liu J, Wang C, Zhang J. Excavation and characterization of key circRNAs for milk fat percentage in Holstein cattle. J Anim Sci 2023; 101:skad157. [PMID: 37209411 PMCID: PMC10290504 DOI: 10.1093/jas/skad157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/19/2023] [Indexed: 05/22/2023] Open
Abstract
Milk fat percentage is one of the significant indicators governing the price and quality of milk and is regulated by a variety of non-coding RNAs. We used RNA sequencing (RNA-seq) techniques and bioinformatics approaches to explore potential candidate circular RNAs (circRNAs) regulating milk fat metabolism. After analysis, compared with low milk fat percentage (LMF) cows, 309 circRNAs were significantly differentially expressed in high milk fat percentage (HMF) cows. Functional enrichment and pathway analysis revealed that the main functions of the parental genes of differentially expressed circRNAs (DE-circRNAs) were related to lipid metabolism. We selected four circRNAs (Novel_circ_0000856, Novel_circ_0011157, novel_circ_0011944, and Novel_circ_0018279) derived from parental genes related to lipid metabolism as key candidate DE-circRNAs. Their head-to-tail splicing was demonstrated by linear RNase R digestion experiments and Sanger sequencing. However, the tissue expression profiles showed that only Novel_circ_0000856, Novel_circ_0011157, and Novel_circ_0011944 were expressed with high abundance in breast tissue. Based on the subcellular localization found that Novel_circ_0000856, Novel_circ_0011157, and Novel_circ_0011944 mainly function as competitive endogenous RNAs (ceRNAs) in the cytoplasm. Therefore, we constructed their ceRNA regulatory networks, and the five hub target genes (CSF1, TET2, VDR, CD34, and MECP2) in ceRNAs were obtained by CytoHubba and MCODE plugins in Cytoscape, as well as tissue expression profiles analysis of target genes. These genes play a key role as important target genes in lipid metabolism, energy metabolism, and cellular autophagy. The Novel_circ_0000856, Novel_circ_0011157, and Novel_circ_0011944 regulate the expression of hub target genes through interaction with miRNAs and constitute key regulatory networks that may be involved in milk fat metabolism. The circRNAs obtained in this study may act as miRNA sponges and thus influence mammary gland development and lipid metabolism in cows, which improves our understanding of the role of circRNAs in cow lactation.
Collapse
Affiliation(s)
- Xiaofang Feng
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Zhengyun Cai
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Yaling Gu
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Tong Mu
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Baojun Yu
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Ruoshuang Ma
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Jiaming Liu
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Chuanchuan Wang
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Juan Zhang
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
10
|
Feng X, Cai Z, Mu T, Yu B, Wang Y, Ma R, Liu J, Wang C, Zhang J, Gu Y. CircRNA screening and ceRNA network construction for milk fat metabolism in dairy cows. Front Vet Sci 2022; 9:995629. [PMID: 36439356 PMCID: PMC9684208 DOI: 10.3389/fvets.2022.995629] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Milk fat is one of the main reference elements for evaluating milk quality and is a primary objective trait in dairy cattle breeding. In recent years, circular RNAs (circRNAs) have been found to play crucial roles in many biological processes. However, the function and expression profiles of circRNAs in milk fat synthesis in cows are not completely understood. We performed RNA sequencing to analyze the genome-wide expression of circRNA transcripts in bovine mammary epithelial cells (BMECs) from cows with extreme differences in milk fat percentage. We identified candidate differential circRNAs associated with milk fat metabolism using functional enrichment analysis and constructed a lipid metabolism-related competing endogenous RNA (ceRNA) interactive regulatory network. RESULTS A total of 290 circRNAs were significantly differentially expressed (DE-circRNAs) in high milk fat percentage (HMF) cows compared to that in low milk fat percentage (LMF) cows. Of the 290 circRNAs, 142 were significantly upregulated and 148 were significantly downregulated. Enrichment analysis (Gene Ontology and Kyoto Encyclopedia of Genes and Genomes) identified four DE-circRNAs (circ_0001122, circ_0007367, circ_0018269, and circ_0015179) that potentially regulate milk fat metabolism. Among them, circ_0001122, circ_0007367, and circ_0015179 had relatively high expression levels in cow mammary gland tissue compared to other tissues (heart, liver, kidney, uterus, ovaries, and small intestine) of cows. The regulatory networks circ_0001122:miR-12043:LIPG, circ_0007367:miR-331-3p:CIDEA/PML, and circ_0018269:miR-11989:RORC/HPX are potential networks to explore the mechanism of milk fat regulation. CONCLUSIONS These results reveal the possible role of circRNAs in milk fat metabolism in dairy cows. Several important circRNAs and ceRNAs affecting milk fat synthesis were identified, providing insights into the complex biology of milk fat synthesis as well as a novel theoretical perspective for future research on lactation, milk quality, and breed improvement in dairy cows.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaling Gu
- Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| |
Collapse
|
11
|
The Mammary Gland: Basic Structure and Molecular Signaling during Development. Int J Mol Sci 2022; 23:ijms23073883. [PMID: 35409243 PMCID: PMC8998991 DOI: 10.3390/ijms23073883] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
The mammary gland is a compound, branched tubuloalveolar structure and a major characteristic of mammals. The mammary gland has evolved from epidermal apocrine glands, the skin glands as an accessory reproductive organ to support postnatal survival of offspring by producing milk as a source of nutrition. The mammary gland development begins during embryogenesis as a rudimentary structure that grows into an elementary branched ductal tree and is embedded in one end of a larger mammary fat pad at birth. At the onset of ovarian function at puberty, the rudimentary ductal system undergoes dramatic morphogenetic change with ductal elongation and branching. During pregnancy, the alveolar differentiation and tertiary branching are completed, and during lactation, the mature milk-producing glands eventually develop. The early stages of mammary development are hormonal independent, whereas during puberty and pregnancy, mammary gland development is hormonal dependent. We highlight the current understanding of molecular regulators involved during different stages of mammary gland development.
Collapse
|
12
|
Rutherford TR, Elder AM, Lyons TR. Anoikis resistance in mammary epithelial cells is mediated by semaphorin 7a. Cell Death Dis 2021; 12:872. [PMID: 34561423 PMCID: PMC8463677 DOI: 10.1038/s41419-021-04133-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022]
Abstract
Semaphorin-7a (SEMA7A), best known as a neuroimmune molecule, plays a diverse role in many cellular processes and pathologies. Here, we show that SEMA7A promotes anoikis resistance in cultured mammary epithelial cells through integrins and activation of pro-survival kinase AKT, which led us to investigate a role for SEMA7A during postpartum mammary gland involution-a normal developmental process where cells die by anoikis. Our results reveal that SEMA7A is expressed on live mammary epithelial cells during involution, that SEMA7A expression is primarily observed in α6-integrin expressing cells, and that luminal progenitor cells, specifically, are decreased in mammary glands of SEMA7A-/- mice during involution. We further identify a SEMA7A-α6/β1-integrin dependent mechanism of mammosphere formation and chemoresistance in mammary epithelial cells and suggest that this mechanism is relevant for recurrence in breast cancer patients.
Collapse
Affiliation(s)
- Taylor R Rutherford
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cell biology, Stem cell, and Development Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alan M Elder
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cancer biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Cell biology, Stem cell, and Development Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Cancer biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- University of Colorado Cancer Center, Aurora, CO, USA.
| |
Collapse
|
13
|
Romagnoli M, Bresson L, Di-Cicco A, Pérez-Lanzón M, Legoix P, Baulande S, de la Grange P, De Arcangelis A, Georges-Labouesse E, Sonnenberg A, Deugnier MA, Glukhova MA, Faraldo MM. Laminin-binding integrins are essential for the maintenance of functional mammary secretory epithelium in lactation. Development 2020; 147:dev.181552. [DOI: 10.1242/dev.181552] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/16/2020] [Indexed: 02/02/2023]
Abstract
Integrin dimers α3/β1, α6/β1 and α6/β4 are the mammary epithelial cell receptors for laminins, which are major components of the mammary basement membrane. The roles of specific basement membrane components and their integrin receptors in the regulation of functional gland development have not been analyzed in detail. To investigate the functions of laminin-binding integrins, we obtained mutant mice with mammary luminal cell-specific deficiencies of the α3 and α6 integrin chains generated by the Cre-Lox approach. During pregnancy, mutant mice displayed decreased luminal progenitor activity and retarded lobulo-alveolar development. Mammary glands appeared functional at the onset of lactation in mutant mice, however myoepithelial cell morphology was markedly altered, suggesting cellular compensation mechanisms involving cytoskeleton reorganization. Notably, lactation was not sustained in mutant females, and the glands underwent precocious involution. Inactivation of the p53 gene rescued the growth defects but did not restore lactogenesis in mutant mice. These results suggest that the p53 pathway is involved in the control of mammary cell proliferation and survival downstream of laminin-binding integrins and underline an essential role of cell interactions with laminin for lactogenic differentiation.
Collapse
Affiliation(s)
- Mathilde Romagnoli
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
| | - Laura Bresson
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
| | - Amandine Di-Cicco
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
| | - María Pérez-Lanzón
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
| | - Patricia Legoix
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | | | - Adèle De Arcangelis
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104/INSERM U964/ULP, F-67404 Illkirch, France
| | - Elisabeth Georges-Labouesse
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104/INSERM U964/ULP, F-67404 Illkirch, France
| | - Arnoud Sonnenberg
- Division of Cell Biology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Marie-Ange Deugnier
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
- Inserm, Paris, F-75013, Paris, France
| | - Marina A. Glukhova
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
- Inserm, Paris, F-75013, Paris, France
| | - Marisa M. Faraldo
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F-75005 Paris, France
- Inserm, Paris, F-75013, Paris, France
| |
Collapse
|
14
|
|
15
|
Zhao X, Ponchon B, Lanctôt S, Lacasse P. Invited review: Accelerating mammary gland involution after drying-off in dairy cattle. J Dairy Sci 2019; 102:6701-6717. [PMID: 31202662 DOI: 10.3168/jds.2019-16377] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/22/2019] [Indexed: 01/20/2023]
Abstract
Bovine mammary gland involution, as a part of the reproductive cycle in dairy cows, is a very important remodeling transformation of the mammary gland for the subsequent lactation. There is considerable incentive to accelerate mammary gland involution to improve udder health, shorten the dry period, and simplify the management process by reducing dietary changes. The complex process of mammary involution is characterized by morphological changes in the epithelial cells and mammary tissue, changes in the composition of mammary secretions, and changes in the integrity of tight junctions. Involution is facilitated by elements of the immune system and several types of proteases and is coordinated by various types of hormones. This review first describes the involution process and then argues for the need to accelerate it. Last, this review focuses on various intervention methods for accelerating involution. Our aim is to provide a comprehensive overview of bovine mammary gland involution as well as potential techniques and new opinions for dry cow management.
Collapse
Affiliation(s)
- X Zhao
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9.
| | - B Ponchon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9
| | - S Lanctôt
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| | - P Lacasse
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| |
Collapse
|
16
|
Jena MK, Jaswal S, Kumar S, Mohanty AK. Molecular mechanism of mammary gland involution: An update. Dev Biol 2019; 445:145-155. [DOI: 10.1016/j.ydbio.2018.11.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/01/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022]
|
17
|
Pišlar A, Jewett A, Kos J. Cysteine cathepsins: Their biological and molecular significance in cancer stem cells. Semin Cancer Biol 2018; 53:168-177. [DOI: 10.1016/j.semcancer.2018.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
|
18
|
Basement membrane extract attenuates the more malignant gene expression profile accentuated by fibronectin in prostate cancer cells. Mol Cell Biochem 2018; 451:131-138. [PMID: 29961211 DOI: 10.1007/s11010-018-3399-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/26/2018] [Indexed: 01/14/2023]
Abstract
Prostate cancer (PCa) has high mortality rates, with most of the deaths resulting from the development of metastasis. Fibronectin (FN) plays key roles in cell adhesion and affects the migratory behavior of cells. In the tumor microenvironment and also in the blood plasma during metastasis, FN displays increased expression, however its role in prostate cancer remains poorly understood. This study aimed to unveil the specific roles of FN as a soluble component, alone or in combination with a complex basement membrane. To investigate the impact of FN in neoplastic prostate cells, we evaluated the gene expression of LNCaP cells by RT-qPCR after exposure to soluble FN (25 µg/mL) either alone or in combination with a basement membrane. When FN was the predominant matrix element, such as in blood plasma, PCa tumor cells increased their expression of genes related to an invasive behavior and resistance to apoptosis, including CDH2, ITGA5, AKT1, and BCL2. However, the combined presence of FN and a complex basement membrane had the opposite effect on LNCaP cells, in which the expression levels of CDH2, ITGA5, AKT1, and BCL2 were reduced. Hierarchical clustering analysis with LNCaP and RWPE-1 cells showed that LNCaP cells exposed to an enriched extracellular matrix displayed an expression pattern more similar to that shown by RWPE-1 cells, a cell line that illustrates characteristics of the normal prostate epithelium. These findings provide the groundwork for future studies addressing the role of FN in tumor growth, particularly in the context of cancer evolution/progression from a solid primary tumor to a transitory circulating state.
Collapse
|
19
|
Miles LA, Baik N, Bai H, Makarenkova HP, Kiosses WB, Krajewski S, Castellino FJ, Valenzuela A, Varki NM, Mueller BM, Parmer RJ. The plasminogen receptor , Plg-R KT, is essential for mammary lobuloalveolar development and lactation. J Thromb Haemost 2018; 16:919-932. [PMID: 29495105 PMCID: PMC5965281 DOI: 10.1111/jth.13988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Indexed: 12/11/2022]
Abstract
Essentials Plg-RKT-/- female mice give birth, but no offspring of Plg-RKT-/- female mice survive to weaning. Causal mechanisms of potential lactational failure in Plg-RKT-/- mice are unknown. Plg-RKT regulates extracellular matrix remodeling, cell proliferation, apoptosis, fibrin surveillance. Plg-RKT is essential for lactogenesis and mammary lobuloalveolar development. SUMMARY Background Lactational competence requires plasminogen, the zymogen of the serine protease, plasmin. Plg-RKT is a unique transmembrane plasminogen receptor that promotes plasminogen activation to plasmin on cell surfaces. Plg-RKT-/- mice are viable, but no offspring of Plg-RKT-/- female mice survive to weaning. Objectives We investigated potential lactational failure in Plg-RKT-/- mice and addressed causal mechanisms. Methods Fibrin accumulation, macrophage infiltration, processing of extracellular matrix components, effects of genetic deletion of fibrinogen, expression of fibrosis genes, and proliferation and apoptosis of epithelial cells were examined in lactating mammary glands of Plg-RKT-/- and Plg-RKT+/+ mice. Results Milk was not present in the stomachs of offspring of Plg-RKT-/- female mice and the pups were rescued by foster mothers. Although the mammary ductal tree developed normally in Plg-RKT-/- glands, lobuloalveolar development was blocked by a hypertrophic fibrotic stroma and infiltrating macrophages were present. A massive accumulation of fibrin was also present in Plg-RKT-/- alveoli and ducts. Although this accumulation was decreased when Plg-RKT-/- mice were made genetically heterozygous for fibrinogen, defects in lobuloalveolar development were not rescued by fibrinogen heterozygosity. Transcriptional profiling revealed that EGF was downregulated 12-fold in Plg-RKT-/- glands. Furthermore, proliferation of epithelial cells was not detectable. In addition, the pro-survival protein, Mcl-1, was markedly downregulated and apoptosis was observed in Plg-RKT-/- but not Plg-RKT+/+ glands. Conclusions Plg-RKT is essential for lactogenesis and functions to maintain the appropriate stromal extracellular matrix environment, regulate epithelial cell proliferation and apoptosis, and, by regulating fibrinolysis, preserve alveolar and ductal patency.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Hongdong Bai
- Veterans Administration San Diego Healthcare System, San Diego, CA
| | | | - William B. Kiosses
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Stan Krajewski
- Sanford-Burnham Medical Research Institute, La Jolla, CA
| | | | - Alex Valenzuela
- Department of Pathology, University of California San Diego, La Jolla, CA
| | - Nissi M. Varki
- Department of Pathology, University of California San Diego, La Jolla, CA
| | | | - Robert J. Parmer
- Veterans Administration San Diego Healthcare System, San Diego, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
20
|
Mettang M, Meyer-Pannwitt V, Karpel-Massler G, Zhou S, Carragher NO, Föhr KJ, Baumann B, Nonnenmacher L, Enzenmüller S, Dahlhaus M, Siegelin MD, Stroh S, Mertens D, Fischer-Posovszky P, Schneider EM, Halatsch ME, Debatin KM, Westhoff MA. Blocking distinct interactions between Glioblastoma cells and their tissue microenvironment: A novel multi-targeted therapeutic approach. Sci Rep 2018; 8:5527. [PMID: 29615749 PMCID: PMC5882900 DOI: 10.1038/s41598-018-23592-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/15/2018] [Indexed: 11/09/2022] Open
Abstract
Due to the highly invasive nature of Glioblastoma (GB), complete surgical resection is not feasible, while motile tumour cells are often associated with several specific brain structures that enhance treatment-resistance. Here, we investigate the therapeutic potential of Disulfiram and Carbenoxolone, that inhibit two distinct interactions between GB and the brain tissue microenvironment: stress-induced cell-matrix adhesion and gap junction mediated cell-cell communication, respectively. Increase in cell numbers of tumour-initiating cells, which are cultured in suspension as cell clusters, and adherent differentiated cells can be blocked to a similar extent by Carbenoxolone, as both cell populations form gap junctions, but the adherent differentiated cells are much more sensitive to Disulfiram treatment, which - via modulation of NF-κB signalling - interferes with cell-substrate adhesion. Interestingly, inducing adhesion in tumour-initiating cells without differentiating them does not sensitize for Disulfiram. Importantly, combining Disulfiram, Carbenoxolone and the standard chemotherapeutic drug Temozolomide reduces tumour size in an orthotopic mouse model. Isolating GB cells from their direct environment within the brain represents an important addition to current therapeutic approaches. The blockage of cellular interactions via the clinically relevant substances Disulfiram and Carbenoxolone, has distinct effects on different cell populations within a tumour, potentially reducing motility and/or resistance to apoptosis.
Collapse
Affiliation(s)
- Melanie Mettang
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.,Institute of Physiological Chemistry, University Medical Center Ulm, Ulm, Germany
| | - Viola Meyer-Pannwitt
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.,Department of Internal Medicine III, University Medical Center Ulm, Ulm, Germany.,Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Shaoxia Zhou
- Department of Clinical Chemistry, University Medical Center Ulm, Ulm, Germany
| | - Neil O Carragher
- Edinburgh Cancer Research Center UK, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Karl Josef Föhr
- Department of Anesthesiology, University Medical Center Ulm, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, University Medical Center Ulm, Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Stefanie Enzenmüller
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Meike Dahlhaus
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sebastien Stroh
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.,Department of Neurology, University Medical Center Ulm, Ulm, Germany
| | - Daniel Mertens
- Department of Internal Medicine III, University Medical Center Ulm, Ulm, Germany.,Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - E Marion Schneider
- Department of Clinical Chemistry, University Medical Center Ulm, Ulm, Germany
| | | | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.
| |
Collapse
|
21
|
Kaylan KB, Gentile SD, Milling LE, Bhinge KN, Kosari F, Underhill GH. Mapping lung tumor cell drug responses as a function of matrix context and genotype using cell microarrays. Integr Biol (Camb) 2017; 8:1221-1231. [PMID: 27796394 DOI: 10.1039/c6ib00179c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Carcinoma progression is influenced by interactions between epithelial tumor cells and components of their microenvironment. In particular, cell-extracellular matrix (ECM) interactions are known to drive tumor growth, metastatic potential, and sensitivity or resistance to therapy. Yet the intrinsic complexity of ECM composition within the tumor microenvironment remains a barrier to comprehensive investigation of these interactions. We present here a high-throughput cell microarray-based approach to study the impact of defined combinations of ECM proteins on tumor cell drug responses. Using this approach, we quantitatively evaluated the effects of 55 different ECM environments representing all single and two-factor combinations of 10 ECM proteins on the responses of lung adenocarcinoma cells to a selection of cancer-relevant small molecule drugs. This drug panel consisted of an alkylating agent and five receptor tyrosine kinase inhibitors. We further determined that expression of the neuroendocrine transcription factor ASCL1, which has been previously associated with poor patient outcome when co-expressed with the RET oncogene, altered cell responses to drugs and modulated cleavage of the pro-apoptotic protein caspase-3 depending on ECM context. Our results suggest that co-expression of specific ECM proteins with known genetic drivers in lung adenocarcinoma may impact therapeutic efficacy. Furthermore, this approach could be utilized to define the molecular mechanisms by which cell-matrix interactions drive drug resistance through integration with clinical cell samples and genomics data.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Stefan D Gentile
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Lauren E Milling
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Kaustubh N Bhinge
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Farhad Kosari
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
22
|
Akhtar N, Li W, Mironov A, Streuli CH. Rac1 Controls Both the Secretory Function of the Mammary Gland and Its Remodeling for Successive Gestations. Dev Cell 2017; 38:522-35. [PMID: 27623383 PMCID: PMC5022528 DOI: 10.1016/j.devcel.2016.08.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 06/30/2016] [Accepted: 08/12/2016] [Indexed: 12/27/2022]
Abstract
An important feature of the mammary gland is its ability to undergo repeated morphological changes during each reproductive cycle with profound tissue expansion in pregnancy and regression in involution. However, the mechanisms that determine the tissue's cyclic regenerative capacity remain elusive. We have now discovered that Cre-Lox ablation of Rac1 in mammary epithelia causes gross enlargement of the epithelial tree and defective alveolar regeneration in a second pregnancy. Architectural defects arise because loss of Rac1 disrupts clearance in involution following the first lactation. We show that Rac1 is crucial for mammary alveolar epithelia to switch from secretion to a phagocytic mode and rapidly remove dying neighbors. Moreover, Rac1 restricts the extrusion of dying cells into the lumen, thus promoting their eradication by live phagocytic neighbors while within the epithelium. Without Rac1, residual milk and cell corpses flood the ductal network, causing gross dilation, chronic inflammation, and defective future regeneration. Rac1 is required for full secretory differentiation of the mammary gland Rac1 restricts apoptotic cell shedding into the lumen to limit inflammation Rac1 contributes to post-lactational tissue remodeling during involution Defective clearance of milk and dead cells in Rac1-null glands causes ductal bloating
Collapse
Affiliation(s)
- Nasreen Akhtar
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK; Department of Oncology and Metabolism, The Bateson Centre, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK.
| | - Weiping Li
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Aleksander Mironov
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Charles H Streuli
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Manchester Breast Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
23
|
Bischof J, Westhoff MA, Wagner JE, Halatsch ME, Trentmann S, Knippschild U, Wirtz CR, Burster T. Cancer stem cells: The potential role of autophagy, proteolysis, and cathepsins in glioblastoma stem cells. Tumour Biol 2017; 39:1010428317692227. [PMID: 28347245 DOI: 10.1177/1010428317692227] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
One major obstacle in cancer therapy is chemoresistance leading to tumor recurrence and metastasis. Cancer stem cells, in particular glioblastoma stem cells, are highly resistant to chemotherapy, radiation, and immune recognition. In case of immune recognition, several survival mechanisms including, regulation of autophagy, proteases, and cell surface major histocompatibility complex class I molecules, are found in glioblastoma stem cells. In different pathways, cathepsins play a crucial role in processing functional proteins that are necessary for several processes and proper cell function. Consequently, strategies targeting these pathways in glioblastoma stem cells are promising approaches to interfere with tumor cell survival and will be discussed in this review.
Collapse
Affiliation(s)
- Joachim Bischof
- 1 Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Mike-Andrew Westhoff
- 2 Department Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Johanna Elisabeth Wagner
- 3 Department of Neurosurgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Marc-Eric Halatsch
- 3 Department of Neurosurgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Stephanie Trentmann
- 1 Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Uwe Knippschild
- 1 Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Christian Rainer Wirtz
- 3 Department of Neurosurgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| | - Timo Burster
- 3 Department of Neurosurgery, Surgery Center, Ulm University Medical Center, Ulm University, Ulm, Germany
| |
Collapse
|
24
|
Moreno-Layseca P, Ucar A, Sun H, Wood A, Olabi S, Gilmore AP, Brennan K, Streuli CH. The requirement of integrins for breast epithelial proliferation. Eur J Cell Biol 2017; 96:227-239. [PMID: 28363396 DOI: 10.1016/j.ejcb.2017.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/24/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022] Open
Abstract
Epithelial cells forming mammary gland ducts and alveoli require adhesion to the extracellular matrix for their function. Mammary epithelial cells need β1-integrins for normal cell cycle regulation. However, the role of β1-integrins in tumorigenesis has not been fully resolved. β1-integrin is necessary for tumour formation in transgenic mice expressing the Polyomavirus Middle T antigen, but it is dispensable in those overexpressing ErbB2. This suggests that some oncogenes can manage without β1-integrin to proliferate and form tumours, while others still require it. Here we have developed a model to test whether expression of an oncogene can surpass the need for β1-integrin to drive proliferation. We co-expressed the ErbB2 or Akt oncogenes with shRNA to target β1-integrin in mammary epithelial cells, and found that they show a differential dependence on β1-integrin for cell division. Moreover, we identified a key proliferative role of the Rac1-Pak axis downstream of β1-integrin signalling. Our data suggest that, in mammary epithelial cells, oncogenes with the ability to signal to Pak surpass the requirement of integrins for malignant transformation. This highlights the importance of using the correct combination therapy for breast cancer, depending on the oncogenes expressed in the tumour.
Collapse
Affiliation(s)
- Paulina Moreno-Layseca
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Ahmet Ucar
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Heyuan Sun
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Amber Wood
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Safiah Olabi
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Andrew P Gilmore
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Keith Brennan
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Charles H Streuli
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| |
Collapse
|
25
|
Yang N, Williams J, Pekovic-Vaughan V, Wang P, Olabi S, McConnell J, Gossan N, Hughes A, Cheung J, Streuli CH, Meng QJ. Cellular mechano-environment regulates the mammary circadian clock. Nat Commun 2017; 8:14287. [PMID: 28134247 PMCID: PMC5290282 DOI: 10.1038/ncomms14287] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Circadian clocks drive ∼24 h rhythms in tissue physiology. They rely on transcriptional/translational feedback loops driven by interacting networks of clock complexes. However, little is known about how cell-intrinsic circadian clocks sense and respond to their microenvironment. Here, we reveal that the breast epithelial clock is regulated by the mechano-chemical stiffness of the cellular microenvironment in primary cell culture. Moreover, the mammary clock is controlled by the periductal extracellular matrix in vivo, which contributes to a dampened circadian rhythm during ageing. Mechanistically, the tension sensing cell-matrix adhesion molecule, vinculin, and the Rho/ROCK pathway, which transduces signals provided by extracellular stiffness into cells, regulate the activity of the core circadian clock complex. We also show that genetic perturbation, or age-associated disruption of self-sustained clocks, compromises the self-renewal capacity of mammary epithelia. Thus, circadian clocks are mechano-sensitive, providing a potential mechanism to explain how ageing influences their amplitude and function.
Collapse
Affiliation(s)
- Nan Yang
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Jack Williams
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Vanja Pekovic-Vaughan
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Pengbo Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Safiah Olabi
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - James McConnell
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nicole Gossan
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Alun Hughes
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Julia Cheung
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Charles H. Streuli
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Qing-Jun Meng
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
26
|
Miles LA, Baik N, Lighvani S, Khaldoyanidi S, Varki NM, Bai H, Mueller B, Parmer RJ. Deficiency of plasminogen receptor, Plg-R KT , causes defects in plasminogen binding and inflammatory macrophage recruitment in vivo. J Thromb Haemost 2017; 15:155-162. [PMID: 27714956 PMCID: PMC5280214 DOI: 10.1111/jth.13532] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/02/2016] [Indexed: 12/11/2022]
Abstract
Essentials Plg-RKT is a novel integral membrane plasminogen receptor. The functions of Plg-RKT in vivo are not known. Plg-RKT is a key player in macrophage recruitment in the inflammatory response in vivo. Plg-RKT deficiency is not compatible with survival of the species. SUMMARY Background Plg-RKT is a novel integral membrane plasminogen receptor that binds plasminogen via a C-terminal lysine exposed on the cell surface and promotes plasminogen activation on the cell surface by both tissue plasminogen activator and urokinase plasminogen activator. Objectives To evaluate the role of Plg-RKT in vivo we generated Plg-RKT-/- mice using a homologous recombination technique. Methods We characterized the effect of Plg-RKT deletion on reproduction, viability, health and spontaneous thrombosis and inflammation. Results Plg-RKT-/- mice were viable and fertile. Survival of Plg-RKT-/- mice and Plg-RKT+/+ littermates was not significantly different. However, quite strikingly, all pups of Plg-RKT-/- females died within 2 days of birth, consistent with a lactation defect in Plg-RKT-/- mothers. Additionally, there was a significant effect of Plg-RKT deficiency on the growth rates of female, but not male, mice. In experimental peritonitis studies, Plg-RKT-/- mice exhibited a marked defect in macrophage recruitment. As a contributing mechanism, the capacity of Plg-RKT-/- macrophages for plasminogen binding was markedly decreased. Conclusions These studies demonstrate that Plg-RKT is required for plasminogen binding and macrophage migration in vivo. In addition, Plg-RKT deficiency is not compatible with survival of the species, due to the death of all offspring of Plg-RKT-/- females. This new mouse model will be important for future studies aimed at delineating the role of cell surface plasminogen activation in challenge and disease models in vivo.
Collapse
Affiliation(s)
- L. A. Miles
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| | - N. Baik
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| | - S. Lighvani
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| | - S. Khaldoyanidi
- Department of Regenerative Medicine, Torrey Pines Institute for Molecular Studies, La Jolla, CA
| | - N. M. Varki
- Department of Pathology, University of California San Diego, La Jolla, CA
| | - H. Bai
- Veterans Administration San Diego Healthcare System, San Diego, CA
| | - B.M. Mueller
- San Diego Biomedical Research Institute, San Diego, CA
| | - R. J. Parmer
- Veterans Administration San Diego Healthcare System, San Diego, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
27
|
Maya-Mendoza A, Bartek J, Jackson DA, Streuli CH. Cellular microenvironment controls the nuclear architecture of breast epithelia through β1-integrin. Cell Cycle 2016; 15:345-56. [PMID: 26818565 PMCID: PMC4943696 DOI: 10.1080/15384101.2015.1121354] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Defects in nuclear architecture occur in a variety of diseases, however the fundamental mechanisms that control the internal structure of nuclei are poorly defined. Here we reveal that the cellular microenvironment has a profound influence on the global internal organization of nuclei in breast epithelia. A 3D microenvironment induces a prolonged but reversible form of cell cycle arrest that features many of the classical markers of cell senescence. This unique form of arrest is dependent on signaling from the external microenvironment through β1-integrins. It is concomitant with alterations in nuclear architecture that characterize the withdrawal from cell proliferation. Unexpectedly, following prolonged cell cycle arrest in 3D, the senescence-like state and associated reprogramming of nuclear architecture are freely reversible on altering the dimensionality of the cellular microenvironment. Breast epithelia can therefore maintain a proliferative plasticity that correlates with nuclear remodelling. However, the changes in nuclear architecture are cell lineage-specific and do not occur in fibroblasts, and moreover they are overcome in breast cancer cells.
Collapse
Affiliation(s)
- Apolinar Maya-Mendoza
- a Faculty of Life Sciences and Wellcome Trust Center for Cell-Matrix Research, University of Manchester , Manchester , United Kingdom.,b Department of Genome Integrity , Danish Cancer Society Research Center , Copenhagen , Denmark
| | - Jiri Bartek
- b Department of Genome Integrity , Danish Cancer Society Research Center , Copenhagen , Denmark.,c Science for Life Laboratory, Division of Translational Medicine and Chemical Biology , Department of Medical Biochemistry and Biophysics, Karolinska Institute , Stockholm , Sweden
| | - Dean A Jackson
- a Faculty of Life Sciences and Wellcome Trust Center for Cell-Matrix Research, University of Manchester , Manchester , United Kingdom
| | - Charles H Streuli
- a Faculty of Life Sciences and Wellcome Trust Center for Cell-Matrix Research, University of Manchester , Manchester , United Kingdom
| |
Collapse
|
28
|
Čunderlíková B. Clinical significance of immunohistochemically detected extracellular matrix proteins and their spatial distribution in primary cancer. Crit Rev Oncol Hematol 2016; 105:127-44. [DOI: 10.1016/j.critrevonc.2016.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 04/03/2016] [Accepted: 04/27/2016] [Indexed: 02/07/2023] Open
|
29
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
30
|
Loss of anchorage primarily induces non-apoptotic cell death in a human mammary epithelial cell line under atypical focal adhesion kinase signaling. Cell Death Dis 2015; 6:e1619. [PMID: 25611393 PMCID: PMC4669778 DOI: 10.1038/cddis.2014.583] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 12/05/2014] [Accepted: 12/10/2014] [Indexed: 12/27/2022]
Abstract
Anchorage dependence of cellular growth and survival prevents inappropriate cell growth or survival in ectopic environments, and serves as a potential barrier to metastasis of cancer cells. Therefore, obtaining a better understanding of anchorage-dependent responses in normal cells is the first step to understand and impede anchorage independence of growth and survival in cancer cells and finally to eradicate cancer cells during metastasis. Anoikis, a type of apoptosis specifically induced by lack of appropriate cell-extracellular matrix adhesion, has been established as the dominant response of normal epithelial cells to anchorage loss. For example, under detached conditions, the untransformed mammary epithelial cell (MEC) line MCF-10 A, which exhibits myoepithelial characteristics, underwent anoikis dependent on classical ERK signaling. On the other hand, recent studies have revealed a variety of phenotypes resulting in cell death modalities distinct from anoikis, such as autophagy, necrosis, and cornification, in detached epithelial cells. In the present study, we characterized detachment-induced cell death (DICD) in primary human MECs immortalized with hTERT (TertHMECs), which are bipotent progenitor-like cells with a differentiating phenotype to luminal cells. In contrast to MCF-10 A cells, apoptosis was not observed in detached TertHMECs; instead, non-apoptotic cell death marked by features of entosis, cornification, and necrosis was observed along with downregulation of focal adhesion kinase (FAK) signaling. Cell death was overcome by anchorage-independent activities of FAK but not PI3K/AKT, SRC, and MEK/ERK, suggesting critical roles of atypical FAK signaling pathways in the regulation of non-apoptotic cell death. Further analysis revealed an important role of TRAIL (tumor necrosis factor (TNF)-related apoptosis-inducing ligand) as a mediator of FAK signaling in regulation of entosis and necrosis and a role of p38 MAPK in the induction of necrosis. Overall, the present study highlighted outstanding cell subtype or differentiation stage specificity in cell death phenotypes induced upon anchorage loss in human MECs.
Collapse
|
31
|
Engel N, Falodun A, Kühn J, Kragl U, Langer P, Nebe B. Pro-apoptotic and anti-adhesive effects of four African plant extracts on the breast cancer cell line MCF-7. Altern Ther Health Med 2014; 14:334. [PMID: 25199565 PMCID: PMC4177160 DOI: 10.1186/1472-6882-14-334] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 08/27/2014] [Indexed: 11/18/2022]
Abstract
Background Jatropha curcas (JCP1), Pyrenacantha staudtii (PS), Picralima nitida (ZI) and Jatropha gossypifolia (JCP2) are plants used in the African folklore for the treatment of various cancers. Methods This study investigated the in vitro anticancer effects of the ethanol extracts against human epithelial MCF-7 breast cancer cells in a dose-dependent manner (1–50 μg/ml) by using cell cycle analysis, viability assay, annexin V/PI staining, TUNEL method and expression determination of apoptotic and adhesion relevant proteins. Adhesion processes were monitored by detachment via flow cytometry, β1-integrin expression and formation of the actin cytoskeleton. Results The three extracts, termed PS, JCP1 and JCP2 at a concentration of 10 μg/ml induced cell death in MCF-7 breast cancer cells verified by high amounts of PI-positive cells in the cell cycle analysis, Annexin V/PI staining and DNA fragmentation measurements. In parallel cell detachment was accompanied by decreased β1- integrin expression and phosphorylation of the focal adhesion kinase at Tyr397. ZI extract was the exception by the increasing β1-integrin expression and strengthening the cortical actin cytoskeleton. However, all four plant extracts mediated strong anti-cancer properties with IC50 values between 23–38 μg/ml. Conclusion PS, JCP1 and JCP2 were found to be very active against MCF-7 cells by inducing anoikis and therefore possessing vast potential as medicinal drugs especially in estrogen receptor positive breast cancer treatment. ZI mediated their anti-cancer action by different signaling mechanisms which should be analyzed in future studies. Our results further supported the idea that medicinal plants can be promising sources of putative anticancer agents. Electronic supplementary material The online version of this article (doi:10.1186/1472-6882-14-334) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Lim YZ, South AP. Tumour-stroma crosstalk in the development of squamous cell carcinoma. Int J Biochem Cell Biol 2014; 53:450-8. [PMID: 24955488 DOI: 10.1016/j.biocel.2014.06.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/13/2014] [Accepted: 06/14/2014] [Indexed: 12/23/2022]
Abstract
Squamous cell carcinoma (SCC) represents one of the most frequently diagnosed tumours and contributes significant mortality worldwide. Recent deep sequencing of cancer genomes has identified common mutations in SCC arising across different tissues highlighting perturbation of squamous differentiation as a key event. At the same time significant data have been accumulating to show that common tumour-stroma interactions capable of driving disease progression are also evident when comparing SCC arising in different tissues. We and others have shown altered matrix composition surrounding SCC can promote tumour development. This review focuses on some of the emerging data with particular emphasis on SCC of head and neck and skin with discussion on the potential tumour suppressive properties of a normal microenvironment. Such data indicate that regardless of the extent and type of somatic mutation it is in fact the tumour context that defines metastatic progression.
Collapse
Affiliation(s)
- Yok Zuan Lim
- Division of Cancer Research, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, UK; Institute of Medical Biology, A*Star, Singapore
| | - Andrew P South
- Division of Cancer Research, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, UK; Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, United States.
| |
Collapse
|
33
|
Anderson LR, Owens TW, Naylor MJ. Integrins in development and cancer. Biophys Rev 2014; 6:191-202. [PMID: 28510181 PMCID: PMC5418411 DOI: 10.1007/s12551-013-0123-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/28/2013] [Indexed: 01/13/2023] Open
Abstract
The correct control of cell fate decisions is critical for metazoan development and tissue homeostasis. It is established that the integrin family of cell surface receptors regulate cell fate by mediating cell-cell and cell-extracellular matrix (ECM) interactions. However, our understanding of how the different family members control discrete aspects of cell biology, and how this varies between tissues and is temporally regulated, is still in its infancy. An emerging area of investigation aims to understand how integrins translate changes in tension in the surrounding microenvironment into biological responses. This is particularly pertinent due to changes in the mechanical properties of the ECM having been linked to diseases, such as cancer. In this review, we provide an overview of the roles integrins play in important developmental processes, such as proliferation, polarity, apoptosis, differentiation and maintenance of "stemness". We also discuss recent advances in integrin mechanobiology and highlight the involvement of integrins and aberrant ECM in cancer.
Collapse
Affiliation(s)
- Luke R Anderson
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Thomas W Owens
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Matthew J Naylor
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, Room E212, Anderson Stuart Building (F13), Sydney, NSW, 2006, Australia.
| |
Collapse
|
34
|
Westhoff MA, Brühl O, Nonnenmacher L, Karpel-Massler G, Debatin KM. Killing me softly--future challenges in apoptosis research. Int J Mol Sci 2014; 15:3746-67. [PMID: 24595238 PMCID: PMC3975365 DOI: 10.3390/ijms15033746] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/10/2014] [Accepted: 02/19/2014] [Indexed: 12/28/2022] Open
Abstract
The induction of apoptosis, a highly regulated and clearly defined mode of cell dying, is a vital tenet of modern cancer therapy. In this review we focus on three aspects of apoptosis research which we believe are the most crucial and most exciting areas currently investigated and that will need to be better understood in order to enhance the efficacy of therapeutic measures. First, we discuss which target to select for cancer therapy and argue that not the cancer cell as such, but its interaction with the microenvironment is a more promising and genetically stable site of attack. Second, the complexity of combination therapy is elucidated using the PI3-K-mediated signaling network as a specific example. Here we show that the current clinical approach to sensitize malignancies to apoptosis by maximal, prolonged inhibition of so-called survival pathways can actually be counter productive. Third, we propose that under certain conditions which will need to be clearly defined in future, chronification of a tumor might be preferable to the attempt at a cure. Finally, we discuss further problems with utilizing apoptosis induction in cancer therapy and propose a novel potential therapeutic approach that combines the previously discussed features.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm 89075, Germany.
| | - Oliver Brühl
- Laboratorio Analisi Sicilia Catania, Lentini (SR) 96016, Italy.
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm 89075, Germany.
| | | | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm 89075, Germany.
| |
Collapse
|
35
|
Boutinaud M, Galio L, Lollivier V, Finot L, Wiart S, Esquerré D, Devinoy E. Unilateral once daily milking locally induces differential gene expression in both mammary tissue and milk epithelial cells revealing mammary remodeling. Physiol Genomics 2013; 45:973-85. [PMID: 23983197 DOI: 10.1152/physiolgenomics.00059.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Once daily milking reduces milk yield, but the underlying mechanisms are not yet fully understood. Local regulation due to milk stasis in the tissue may contribute to this effect, but such mechanisms have not yet been fully described. To challenge this hypothesis, one udder half of six Holstein dairy cows was milked once a day (ODM), and the other twice a day (TDM). On the 8th day of unilateral ODM, mammary epithelial cells (MEC) were purified from the milk using immunomagnetic separation. Mammary biopsies were harvested from both udder halves. The differences in transcript profiles between biopsies from ODM and TDM udder halves were analyzed by a 22k bovine oligonucleotide array, revealing 490 transcripts that were differentially expressed. The principal category of upregulated transcripts concerned mechanisms involved in cell proliferation and death. We further confirmed remodeling of the mammary tissue by immunohistochemistry, which showed less cell proliferation and more apoptosis in ODM udder halves. Gene expression analyzed by RT-qPCR in MEC purified from milk and mammary biopsies showed a common downregulation of six transcripts (ABCG2, FABP3, NUCB2, RNASE1 and 5, and SLC34A2) but also some discrepancies. First, none of the upregulated transcripts in biopsies varied in milk-purified MEC. Second, only milk-purified MEC showed significant LALBA downregulation, which suggests therefore that they correspond to a mammary epithelial cell subpopulation. Our results, obtained after unilateral milking, suggest that cell remodeling during ODM is due to a local effect, which may be triggered by milk accumulation.
Collapse
Affiliation(s)
- Marion Boutinaud
- INRA, UR1196 Génomique et Physiologie de la Lactation, Jouy-en-Josas, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Tran T, Teoh CM, Tam JKC, Qiao Y, Chin CY, Chong OK, Stewart AG, Harris T, Wong WSF, Guan SP, Leung BP, Gerthoffer WT, Unruh H, Halayko AJ. Laminin drives survival signals to promote a contractile smooth muscle phenotype and airway hyperreactivity. FASEB J 2013; 27:3991-4003. [PMID: 23756649 DOI: 10.1096/fj.12-221341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Increased airway smooth muscle (ASM) mass is believed to underlie the relatively fixed airway hyperresponsiveness (AHR) in asthma. Developments of therapeutic approaches to reverse airway remodeling are impeded by our lack of insight on the mechanisms behind the increase in mass of contractile ASM cells. Increased expression of laminin, an extracellular matrix protein, is associated with asthma. Our studies investigate the role of laminin-induced ASM survival signals in the development of increased ASM and AHR. Antagonizing laminin integrin binding using the laminin-selective competing peptide, YIGSR, and mimicking laminin with exogenous α2-chain laminin, we show that laminin is both necessary and sufficient to induce ASM cell survival, concomitant with the induction of ASM contractile phenotype. Using siRNA, we show that the laminin-binding integrin α7β1 mediates this process. Moreover, in laminin-211-deficient mice, allergen-induced AHR was not observed. Notably, ASM cells from asthmatic airways express a higher abundance of intracellular cell survival proteins, consistent with a role for reduced rates of cell apoptosis in development of ASM hyperplasia. Targeting the laminin-integrin α7β1 signaling pathway may offer new avenues for the development of therapies to reduce the increase in mass of contractile phenotype ASM cells that underlie AHR in asthma.
Collapse
Affiliation(s)
- Thai Tran
- 2Department of Physiology, National University of Singapore, Block MD9, 2 Medical Dr., Singapore 117597. E-Mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Walker TN, Cimakasky LM, Coleman EM, Madison MN, Hildreth JE. Antibody against integrin lymphocyte function-associated antigen 1 inhibits HIV type 1 infection in primary cells through caspase-8-mediated apoptosis. AIDS Res Hum Retroviruses 2013; 29:371-83. [PMID: 22697794 DOI: 10.1089/aid.2011.0395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
HIV-1 infection induces formation of a virological synapse wherein CD4, chemokine receptors, and cell-adhesion molecules such as lymphocyte function-associated antigen 1 (LFA-1) form localized domains on the cell surface. Studies show that LFA-1 on the surface of HIV-1 particles retains its adhesion function and enhances virus attachment to susceptible cells by binding its counterreceptor intercellular adhesion molecule 1 (ICAM-1). This virus-cell interaction augments virus infectivity by facilitating binding and entry events. In this study, we demonstrate that inhibition of the LFA-1/ICAM-1 interaction by a monoclonal antibody leads to decreased virus production and spread in association with increased apoptosis of HIV-infected primary T cells. The data indicate that the LFA-1/ICAM-1 interaction may limit apoptosis in HIV-1-infected T cells. This phenomenon appears similar to anoikis wherein epithelial cells are protected from apoptosis conferred by ligand-bound integrins. These results have implications for further understanding HIV pathogenesis and replication in peripheral compartments and lymphoid organs.
Collapse
Affiliation(s)
- Tiffany N. Walker
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee
| | | | - Ebony M. Coleman
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, California
| | - M. Nia Madison
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee
| | - James E.K. Hildreth
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, California
| |
Collapse
|
38
|
Akhtar N, Streuli CH. An integrin-ILK-microtubule network orients cell polarity and lumen formation in glandular epithelium. Nat Cell Biol 2013; 15:17-27. [PMID: 23263281 PMCID: PMC3701152 DOI: 10.1038/ncb2646] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/07/2012] [Indexed: 12/16/2022]
Abstract
The extracellular matrix has a crucial role in determining the spatial orientation of epithelial polarity and the formation of lumens in glandular tissues; however, the underlying mechanisms remain elusive. By using Cre–Lox deletion we show that β1 integrins are required for normal mammary gland morphogenesis and lumen formation, both in vivo and in a three-dimensional primary culture model in which epithelial cells directly contact a basement membrane. Downstream of basement membrane β1 integrins, Rac1 is not involved; however, ILK is needed to polarize microtubule plus ends at the basolateral membrane and disrupting each of these components prevents lumen formation. The integrin–microtubule axis is necessary for the endocytic removal of apical proteins from the basement-membrane–cell interface and for internal Golgi positioning. We propose that this integrin signalling network controls the delivery of apical components to the correct surface and thereby governs the orientation of polarity and development of lumens.
Collapse
Affiliation(s)
- Nasreen Akhtar
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M139PT, UK.
| | | |
Collapse
|
39
|
Galio L, Droineau S, Yeboah P, Boudiaf H, Bouet S, Truchet S, Devinoy E. MicroRNA in the ovine mammary gland during early pregnancy: spatial and temporal expression of miR-21, miR-205, and miR-200. Physiol Genomics 2012; 45:151-61. [PMID: 23269700 DOI: 10.1152/physiolgenomics.00091.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The mammary gland undergoes extensive remodeling between the beginning of pregnancy and lactation; this involves cellular processes including cell proliferation, differentiation, and apoptosis, all of which are under the control of numerous regulators. To unravel the role played by miRNA, we describe here 47 new ovine miRNA cloned from mammary gland in early pregnancy displaying strong similarities with those already identified in the cow, human, or mouse. A microarray study of miRNA variations in the adult ovine mammary gland during pregnancy and lactation showed that 100 miRNA are regulated according to three principal patterns of expression: a decrease in early pregnancy, a peak at midpregnancy, or an increase throughout late pregnancy and lactation. One miRNA displaying each pattern (miR-21, miR-205, and miR-200b) was analyzed by qRT-PCR. Variations in expression were confirmed for all three miRNA. Using in situ hybridization, we detected both miR-21 and miR-200 in luminal mammary epithelial cells when expressed, whereas miR-205 was expressed in basal cells during the first half of pregnancy and then in luminal cells during the second half. We therefore conclude that miR-21 is strongly expressed in the luminal cells of the normal mammary gland during early pregnancy when extensive cell proliferation occurs. In addition, we show that miR-205 and miR-200 are coexpressed in luminal cells, but only during the second half of pregnancy. These two miRNA may cooperate to maintain epithelial status by repressing an EMT-like program, to achieve and preserve the secretory phenotype of mammary epithelial cells.
Collapse
Affiliation(s)
- Laurent Galio
- Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1196 Génomique et Physiologie de la Lactation, Jouy-en-Josas, France.
| | | | | | | | | | | | | |
Collapse
|
40
|
Bush KT, Crawford BE, Garner OB, Nigam KB, Esko JD, Nigam SK. N-sulfation of heparan sulfate regulates early branching events in the developing mammary gland. J Biol Chem 2012; 287:42064-70. [PMID: 23060443 DOI: 10.1074/jbc.m112.423327] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Branching morphogenesis, a fundamental process in the development of epithelial organs (e.g. breast, kidney, lung, salivary gland, prostate, pancreas), is in part dependent on sulfation of heparan sulfate proteoglycans. Proper sulfation is mediated by biosynthetic enzymes, including exostosin-2 (Ext2), N-deacetylase/N-sulfotransferases and heparan sulfate O-sulfotransferases. Recent conditional knockouts indicate that whereas primary branching is dependent on heparan sulfate, other stages are dependent upon selective addition of N-sulfate and/or 2-O sulfation (Crawford, B .E., Garner, O. B., Bishop, J. R., Zhang, D. Y., Bush, K. T., Nigam, S. K., and Esko, J. D. (2010) PLoS One 5, e10691; Garner, O .B., Bush, K. T., Nigam, S .K., Yamaguchi, Y., Xu, D., Esko, J. D., and Nigam, S. K. (2011) Dev. Biol. 355, 394-403). Here, we analyzed the effect of deleting both Ndst2 and Ndst1. Whereas deletion of Ndst1 has no major effect on primary or secondary branching, deletion of Ndst2 appears to result in a mild increase in branching. When both genes were deleted, ductal growth was variably diminished (likely due to variable Cre-recombinase activity), but an overabundance of branched structures was evident irrespective of the extent of gland growth or postnatal age. "Hyperbranching" is an unusual phenotype. The effects on N-sulfation and growth factor binding were confirmed biochemically. The results indicate that N-sulfation or a factor requiring N-sulfation regulates primary and secondary branching events in the developing mammary gland. Together with previous work, the data indicate that different stages of ductal branching and lobuloalveolar formation are regulated by distinct sets of heparan sulfate biosynthetic enzymes in an appropriate growth factor context.
Collapse
Affiliation(s)
- Kevin T Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Integrin-mediated adhesion to extracellular matrix proteins is required for survival of many cell types. This phenomenon appears to be a mechanism of tumour suppression and to participate in embryogenesis. Here, our current understanding of how integrin-dependent signals prevent apoptosis and implications of anchorage-dependent survival for development, physiology and pathology are discussed.
Collapse
|
42
|
Yart L, Dessauge F, Finot L, Barbey S, Marnet P, Lollivier V. Ovariectomy improves lactation persistency in dairy cows. J Dairy Sci 2012; 95:3794-802. [DOI: 10.3168/jds.2011-5195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 02/27/2012] [Indexed: 11/19/2022]
|
43
|
Jeanes AI, Wang P, Moreno-Layseca P, Paul N, Cheung J, Tsang R, Akhtar N, Foster FM, Brennan K, Streuli CH. Specific β-containing integrins exert differential control on proliferation and two-dimensional collective cell migration in mammary epithelial cells. J Biol Chem 2012; 287:24103-12. [PMID: 22511753 PMCID: PMC3397837 DOI: 10.1074/jbc.m112.360834] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Understanding how cell cycle is regulated in normal mammary epithelia is essential for deciphering defects of breast cancer and therefore for developing new therapies. Signals provided by both the extracellular matrix and growth factors are essential for epithelial cell proliferation. However, the mechanisms by which adhesion controls cell cycle in normal epithelia are poorly established. In this study, we describe the consequences of removing the β1-integrin gene from primary cultures of mammary epithelial cells in situ, using CreER. Upon β1-integrin gene deletion, the cells were unable to progress efficiently through S-phase, but were still able to undergo collective two-dimensional migration. These responses are explained by the presence of β3-integrin in β1-integrin-null cells, indicating that integrins containing different β-subunits exert differential control on mammary epithelial proliferation and migration. β1-Integrin deletion did not inhibit growth factor signaling to Erk or prevent the recruitment of core adhesome components to focal adhesions. Instead the S-phase arrest resulted from defective Rac activation and Erk translocation to the nucleus. Rac inhibition prevented Erk translocation and blocked proliferation. Activated Rac1 rescued the proliferation defect in β1-integrin-depleted cells, indicating that this GTPase is essential in propagating proliferative β1-integrin signals. These results show that β1-integrins promote cell cycle in mammary epithelial cells, whereas β3-integrins are involved in migration.
Collapse
Affiliation(s)
- Alexa I Jeanes
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Colitti M. BCL-2 Family of Proteins and Mammary Cellular Fate. Anat Histol Embryol 2012; 41:237-47. [DOI: 10.1111/j.1439-0264.2012.01134.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 12/15/2011] [Indexed: 11/29/2022]
Affiliation(s)
- M. Colitti
- Department of Agriculture and Environmental Sciences, Faculty of Veterinary Medicine; University of Udine, via delle Scienze; 206 - 33100; Udine; Italy
| |
Collapse
|
45
|
Garner OB, Bush KT, Nigam KB, Yamaguchi Y, Xu D, Esko JD, Nigam SK. Stage-dependent regulation of mammary ductal branching by heparan sulfate and HGF-cMet signaling. Dev Biol 2011; 355:394-403. [PMID: 21586278 PMCID: PMC3118867 DOI: 10.1016/j.ydbio.2011.04.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/18/2011] [Accepted: 04/30/2011] [Indexed: 02/08/2023]
Abstract
Specific interactions of growth factors with heparan sulfate may function as "switches" to regulate stages of branching morphogenesis in developing mammalian organs, such as breast, lung, salivary gland and kidney, but the evidence derives mostly from studies of explanted tissues or cell culture (Shah et al., 2004). We recently provided in vivo evidence that inactivation of Ndst1, the predominant N-deacetylase/N-sulfotransferase gene essential for the formation of mature heparan sulfate, results in a highly specific defect in murine lobuloalveolar development (Crawford et al., 2010). Here, we demonstrate a highly penetrant dramatic defect in primary branching by mammary epithelial-specific inactivation of Ext1, a subunit of the copolymerase complex that catalyzes the formation of the heparan sulfate chain. In contrast to Ext1 deletion, inactivation of Hs2st (which encodes an enzyme required for 2-O-sulfation of uronic acids in heparan sulfate) did not inhibit ductal formation but displayed markedly decreased secondary and ductal side-branches as well as fewer bifurcated terminal end buds. Targeted conditional deletion of c-Met, the receptor for HGF, in mammary epithelial cells showed similar defects in secondary and ductal side-branching, but did not result in any apparent defect in bifurcation of terminal end buds. Although there is published evidence indicating a role for 2-O sulfation in HGF binding, primary epithelial cells isolated from Hs2st conditional deletions were able to activate Erk in the presence of HGF and there appeared to be only a slight reduction in HGF-mediated c-Met phosphorylation in these cells compared to control. Thus, both c-Met and Hs2st play important, but partly independent, roles in secondary and ductal side-branching. When considered together with previous studies of Ndst1-deficient glands, the data presented here raise the possibility of partially-independent regulation by heparan sulfate-dependent pathways of primary ductal branching, terminal end bud bifurcation, secondary branching, ductal side-branching and lobuloalveolar formation.
Collapse
Affiliation(s)
- Omai B. Garner
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, 92093
| | - Kevin T. Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
| | - Kabir B. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
| | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, 92093
| | - Ding Xu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Sanjay K. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
- Department of Bioengineering, University of California, San Diego, La Jolla, California, 92093
| |
Collapse
|
46
|
Wang Y, Dai Y, Li X, Chen CY, Li W, Yu Z. Inhibition of Smad signaling is implicated in cleft palate induced by all-trans retinoic acid. ACTA BIOLOGICA HUNGARICA 2011; 62:142-50. [PMID: 21555266 DOI: 10.1556/abiol.62.2011.2.4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effect of all-trans retinoic acid (atRA) on palatal fusion and the underlying mechanisms were investigated using organ culture. Compared with control group, the atRA-treated group (1 μM and 5 μM) had more medial edge epithelium (ME) remaining within the midline epithelial seam (MES). At 10 μM atRA, the opposing shelves were not in contact at the culture end (72 h). Cell death detection by TUNEL and laminin immunohistochemistry demonstrated that atRA (5 μM) induced apoptosis in mesenchyme and inhibited degradation of basal lamina within MES. Notably, migration and apoptosis of ME cells and degradation of basal lamina within MES markedly represented vehicle control palatal shelves in culture. Additionally, apoptosis was not detected in mesenchyme of control palatal shelves. Immunoblotting analysis revealed that Smad2 and Smad3 were endogenously activated and expression of Smad7 was inhibited during the fusion process. In contrast, atRA treatment abrogated phosphorylation of Smad2 and Smad3 and inducible expression of Smad7 in ME. From these data, it is assumed that inhibition of Smad pathway by atRA in ME may play a critical role in abrogation of the ME cell apoptosis and degradation of the basal laminin, which might contribute to failure of palatal fusion.
Collapse
Affiliation(s)
- Yuming Wang
- Henan Academy of Medical Sciences, Zhengzhou, China
| | | | | | | | | | | |
Collapse
|
47
|
Jeanes AI, Maya-Mendoza A, Streuli CH. Cellular microenvironment influences the ability of mammary epithelia to undergo cell cycle. PLoS One 2011; 6:e18144. [PMID: 21479230 PMCID: PMC3066216 DOI: 10.1371/journal.pone.0018144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 02/25/2011] [Indexed: 12/11/2022] Open
Abstract
The use of cell culture models is a principal and fundamental technology used in understanding how mammalian cells work. However, for some cell types such as mammary epithelia, the lines selected for extended culture are often transformed or have chromosomal abnormalities, while primary cultures have such a curtailed lifespan that their use is restricted. For example, mammary luminal epithelial cells (MECs) are used to study mechanisms of breast cancer, but the proliferation of primary cell cultures is highly limited. Here we describe the establishment of a new culture system to allow extended analysis of cultures of primary mouse MECs. In 2D monolayer culture, primary MECs showed a burst of proliferation 2-3 days post isolation, after which cell cycle decreased substantially. Addition of mammary epithelial growth factors, such as Epidermal Growth Factor, Fibroblast Growth Factor-2, Hepatocyte Growth Factor, and Receptor Activator for Nuclear Factor κB Ligand, or extracellular matrix proteins did not maintain their proliferation potential, neither did replating the cells to increase the mitogenic response. However, culturing MECs directly after tissue extraction in a 3D microenvironment consisting of basement membrane proteins, extended the time in culture in which the cells could proliferate. Our data reveal that the cellular microenvironment has profound effects on the proliferative properties of the mammary epithelia and is dominant over growth factors. Moreover, manipulating the cellular environment using this novel method can maintain the proliferative potential of primary MECs, thus enabling cell cycle to be studied as an endpoint after gene transfer or gene deletion experiments.
Collapse
Affiliation(s)
- Alexa I. Jeanes
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences,
University of Manchester, Manchester, United Kingdom
| | - Apolinar Maya-Mendoza
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences,
University of Manchester, Manchester, United Kingdom
| | - Charles H. Streuli
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences,
University of Manchester, Manchester, United Kingdom
| |
Collapse
|
48
|
Kohn EA, Du Z, Sato M, Van Schyndle CMH, Welsh MA, Yang YA, Stuelten CH, Tang B, Ju W, Bottinger EP, Wakefield LM. A novel approach for the generation of genetically modified mammary epithelial cell cultures yields new insights into TGFβ signaling in the mammary gland. Breast Cancer Res 2010; 12:R83. [PMID: 20942910 PMCID: PMC3096976 DOI: 10.1186/bcr2728] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 07/29/2010] [Accepted: 10/13/2010] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Molecular dissection of the signaling pathways that underlie complex biological responses in the mammary epithelium is limited by the difficulty of propagating large numbers of mouse mammary epithelial cells, and by the inability of ribonucleic acid interference (RNAi)-based knockdown approaches to fully ablate gene function. Here we describe a method for the generation of conditionally immortalized mammary epithelial cells with defined genetic defects, and we show how such cells can be used to investigate complex signal transduction processes using the transforming growth factor beta (TGFβ/Smad pathway as an example. METHODS We intercrossed the previously described H-2Kb-tsA58 transgenic mouse (Immortomouse) which expresses a temperature-sensitive mutant of the simian virus-40 large T-antigen (tsTAg), with mice of differing Smad genotypes. A panel of conditionally immortalized mammary epithelial cell (IMEC) cultures were derived from the virgin mammary glands of offspring of these crosses and used to assess the Smad dependency of different biological responses to TGFβ. RESULTS IMECs could be propagated indefinitely at permissive temperatures and had a stable epithelial phenotype, resembling primary mammary epithelial cells with respect to several criteria, including responsiveness to TGFβ. Using this panel of cells, we demonstrated that Smad3, but not Smad2, is necessary for TGFβ-induced apoptotic, growth inhibitory and EMT responses, whereas either Smad can support TGFβ-induced invasion as long as a threshold level of total Smad is exceeded. CONCLUSIONS This work demonstrates the practicality and utility of generating conditionally immortalized mammary epithelial cell lines from genetically modified Immortomice for detailed investigation of complex signaling pathways in the mammary epithelium.
Collapse
Affiliation(s)
- Ethan A Kohn
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Zhijun Du
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Misako Sato
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Catherine MH Van Schyndle
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Michael A Welsh
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Yu-an Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Christina H Stuelten
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Binwu Tang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| | - Wenjun Ju
- Department of Internal Medicine, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Erwin P Bottinger
- Division of Nephrology, Department of Medicine, Charles R Bronfman Institute for Personalized Medicine, Mount Sinai School of Medicine, 1468 Madison Avenue, New York, NY 10029, USA
| | - Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, 37 Convent Drive MSC 4255, Bethesda MD 20892, USA
| |
Collapse
|
49
|
Morrison B, Cutler ML. The contribution of adhesion signaling to lactogenesis. J Cell Commun Signal 2010; 4:131-9. [PMID: 21063503 DOI: 10.1007/s12079-010-0099-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 08/30/2010] [Indexed: 11/28/2022] Open
Abstract
The mammary gland undergoes hormonally controlled cycles of pubertal maturation, pregnancy, lactation, and involution, and these processes rely on complex signaling mechanisms, many of which are controlled by cell-cell and cell-matrix adhesion. The adhesion of epithelial cells to the extracellular matrix initiates signaling mechanisms that have an impact on cell proliferation, survival, and differentiation throughout lactation. The control of integrin expression on the mammary epithelial cells, the composition of the extracellular matrix and the presence of secreted matricellular proteins all contribute to essential adhesion signaling during lactogenesis. In vitro and in vivo studies, including the results from genetically engineered mice, have shed light on the regulation of these processes at the cell and tissue level and have led to increased understanding of the essential signaling components that are regulated in temporal and cell specific manner during lactogenesis. Recent studies suggest that a secreted matricellular protein, CTGF/CCN2, may play a role in lactogenic differentiation through binding to β1 integrin complexes, enhancing the production of extracellular matrix components and contributions to cell adhesion signaling.
Collapse
|
50
|
Ramamoorthy S, Dhananjayan SC, Demayo FJ, Nawaz Z. Isoform-specific degradation of PR-B by E6-AP is critical for normal mammary gland development. Mol Endocrinol 2010; 24:2099-113. [PMID: 20829392 DOI: 10.1210/me.2010-0116] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
E6-associated protein (E6-AP), which was originally identified as an ubiquitin-protein ligase, also functions as a coactivator of estrogen (ER-α) and progesterone (PR) receptors. To investigate the in vivo role of E6-AP in mammary gland development, we generated transgenic mouse lines that either overexpress wild-type (WT) human E6-AP (E6-AP(WT)) or ubiquitin-protein ligase-defective E6-AP (E6-AP(C833S)) in the mammary gland. Here we show that overexpression of E6-AP(WT) results in impaired mammary gland development. In contrast, overexpression of E6-AP(C833S) or loss of E6-AP (E6-AP(KO)) increases lateral branching and alveolus-like protuberances in the mammary gland. We also show that the mammary phenotypes observed in the E6-AP transgenic and knockout mice are due, in large part, to the alteration of PR-B protein levels. We also observed alteration in ER-α protein level, which might contribute to the observed mammary phenotype by regulating PR expression. Furthermore, E6-AP regulates PR-B protein levels via the ubiquitin-proteasome pathway. Additionally, we also show that E6-AP impairs progesterone-induced Wnt-4 expression by decreasing the steady state level of PR-B in both mice and in human breast cancer cells. In conclusion, we present the novel observation that E6-AP controls mammary gland development by regulating PR-B protein turnover via the ubiquitin proteasome pathway. For the first time, we show that the E3-ligase activity rather than the coactivation function of E6-AP plays an important role in the mammary gland development, and the ubiquitin-dependent PR-B degradation is not required for its transactivation functions. This mechanism appears to regulate normal mammogenesis, and dysregulation of this process may be an important contributor to mammary cancer development and progression.
Collapse
Affiliation(s)
- Sivapriya Ramamoorthy
- Department of Biochemistry & Molecular Biology, Braman Breast Cancer Institute (M-877), University of Miami School of Medicine, Batchelor Building, Room 416, 1580 Northwest 10 Avenue, Miami, Florida 33136, USA
| | | | | | | |
Collapse
|