1
|
Faktor J, Kote S, Bienkowski M, Hupp TR, Marek-Trzonkowska N. Novel FFPE proteomics method suggests prolactin induced protein as hormone induced cytoskeleton remodeling spatial biomarker. Commun Biol 2024; 7:708. [PMID: 38851810 PMCID: PMC11162451 DOI: 10.1038/s42003-024-06354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/20/2024] [Indexed: 06/10/2024] Open
Abstract
Robotically assisted proteomics provides insights into the regulation of multiple proteins achieving excellent spatial resolution. However, developing an effective method for spatially resolved quantitative proteomics of formalin fixed paraffin embedded tissue (FFPE) in an accessible and economical manner remains challenging. We introduce non-robotic In-insert FFPE proteomics approach, combining glass insert FFPE tissue processing with spatial quantitative data-independent mass spectrometry (DIA). In-insert approach identifies 450 proteins from a 5 µm thick breast FFPE tissue voxel with 50 µm lateral dimensions covering several tens of cells. Furthermore, In-insert approach associated a keratin series and moesin (MOES) with prolactin-induced protein (PIP) indicating their prolactin and/or estrogen regulation. Our data suggest that PIP is a spatial biomarker for hormonally triggered cytoskeletal remodeling, potentially useful for screening hormonally affected hotspots in breast tissue. In-insert proteomics represents an alternative FFPE processing method, requiring minimal laboratory equipment and skills to generate spatial proteotype repositories from FFPE tissue.
Collapse
Affiliation(s)
- Jakub Faktor
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822, Gdansk, Poland.
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822, Gdansk, Poland.
| | - Michal Bienkowski
- Medical University of Gdansk, University of Gdansk, Mariana Smoluchowskiego 17, 80-214, Gdansk, Poland
| | - Ted R Hupp
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822, Gdansk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822, Gdansk, Poland
| |
Collapse
|
2
|
Wang F, van Baal J, Ma L, Gao X, Dijkstra J, Bu D. MRCKα is a novel regulator of prolactin-induced lactogenesis in bovine mammary epithelial cells. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 10:319-328. [PMID: 35891685 PMCID: PMC9304597 DOI: 10.1016/j.aninu.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 01/18/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Myotonic dystrophy-related Cdc42-binding kinase alpha (MRCKα) is an integral component of signaling pathways controlling vital cellular processes, including cytoskeletal reorganization, cell proliferation and cell survival. In this study, we investigated the physiological role of MRCKα in milk protein and fat production in dairy cows, which requires a dynamic and strict organization of the cytoskeletal network in bovine mammary epithelial cells (BMEC). Within a selection of 9 Holstein cows, we found that both mRNA and protein expression of MRCKα in the mammary gland were upregulated during lactation and correlated positively (r > 0.89) with the mRNA and protein levels of β-casein. Similar positive correlations (r > 0.79) were found in a primary culture of BMEC stimulated with prolactin for 24 h. In these cells, silencing of MRCKα decreased basal β-casein, sterol-regulatory element binding protein (SREBP)-1 and cyclin D1 protein level, phosphorylation of mTOR, triglyceride secretion, cell number and viability-while overexpression of MRCKα displayed the reversed effect. Notably, silencing of MRCKα completely prevented the stimulatory action of prolactin on the same parameters. These data demonstrate that MRCKα is a critical mediator of prolactin-induced lactogenesis via stimulation of the mTOR/SREBP1/cyclin D1 signaling pathway.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Jürgen van Baal
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Lu Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, 434020, China
| | - Jan Dijkstra
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Dengpan Bu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Joint Laboratory on Integrated Crop-Tree-Livestock Systems of the Chinese Academy of Agricultural Sciences (CAAS), Ethiopian Institute of Agricultural Research (EIAR) and World Agroforestry Center (ICRAF), Beijing, 100193, China
| |
Collapse
|
3
|
Zhao X, Sun P, Liu M, Liu S, Huo L, Ding Z, Liu M, Wang S, Lv C, Wu H, Yang L, Liang A. Deoxynivalenol exposure inhibits biosynthesis of milk fat and protein by impairing tight junction in bovine mammary epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113504. [PMID: 35447471 DOI: 10.1016/j.ecoenv.2022.113504] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
Deoxynivalenol (DON) is one of the most common feed contaminants, and it poses a serious threat to the health of dairy cows. The existing studies of biological toxicity of DON mainly focus on the proliferation, oxidative stress, and inflammation in bovine mammary epithelial cells, while its toxicity on the biosynthesis of milk components has not been well documented. Hence, we investigated the toxic effects and the underlying mechanism of DON on the bovine mammary alveolar cells (MAC-T). Our results showed that exposure to various concentrations of DON significantly inhibited cell proliferation, induced apoptosis, and altered the cell morphology which was manifested by cell distortion and shrinkage. Moreover, the transepithelial electrical resistance (TEER) values of MAC-T cells exposed to DON were gradually decreased in a time- and concentration- dependent manner, but lactate dehydrogenase (LDH) leakage was significantly increased with the maximum increase of 2.4-fold, indicating the cell membrane and tight junctions were damaged by DON. Importantly, DON significantly reduced the synthesis of β-casein and lipid droplets, along with the significantly decreases of phospho-mTOR, phospho-4EBP1, phospho-JAK2, and phospho-STAT5. Gene expression profiles showed that the expressions of several genes related to lipid synthesis and metabolism were changed, including acyl-CoA synthetase short-chain family member 2 (ACSS2), fatty acid binding protein 3 (FABP3), 3-hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1), and insulin-induced gene 1 (INSIG1). GO and KEGG enrichment analyses revealed that the differentially expressed genes (DEGs) were significantly enriched in ribosome, glutathione metabolism, and lipid biosynthetic process, which play important roles in the toxicological process induced by DON. Taken together, DON affects the proliferation and functional differentiation of MAC-T cells, which might be related to the cell junction disruption and morphological alteration. Our data provide new insights into functional differentiation and transcriptomic alterations of MAC-T cells after DON exposure, which contributes to a comprehensive understanding of DON-induced toxicity mechanism.
Collapse
Affiliation(s)
- Xinzhe Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Peihao Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Mingxiao Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Shuanghang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Lijun Huo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Zhiming Ding
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ming Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Shuai Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ce Lv
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Hanxiao Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Liguo Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Aixin Liang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
4
|
Prolactin: A hormone with diverse functions from mammary gland development to cancer metastasis. Semin Cell Dev Biol 2020; 114:159-170. [PMID: 33109441 DOI: 10.1016/j.semcdb.2020.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/04/2020] [Accepted: 10/11/2020] [Indexed: 01/14/2023]
Abstract
Prolactin has a rich mechanistic set of actions and signaling in order to elicit developmental effects in mammals. Historically, prolactin has been appreciated as an endocrine peptide hormone that is responsible for final, functional mammary gland development and lactation. Multiple signaling pathways impacted upon by the microenvironment contribute to cell function and differentiation. Endocrine, autocrine and paracrine signaling are now apparent in not only mammary development, but also in cancer, and involve multiple cell types including those of the immune system. Multiple ligands agonists are capable of binding to the prolactin receptor, potentially expanding receptor function. Prolactin has an important role not only in tumorigenesis of the breast, but also in a number of hormonally responsive cancers such as prostate, ovarian and endometrial cancer, as well as pancreatic and lung cancer. Although pituitary and extra-pituitary sources of prolactin such as the epithelium are important, stromal sourced prolactin is now also being recognized as an important factor in tumor progression, all of which potentially signal to multiple cell types in the tumor microenvironment. While prolactin has important roles in milk production including calcium and bone homeostasis, in the disease state it can also affect bone homeostasis. Prolactin also impacts metastatic cancer of the breast to modulate the bone microenvironment and promote bone damage. Prolactin has a fascinating contribution in both physiologic and pathologic settings of mammals.
Collapse
|
5
|
Duruksu G, Aciksari A. Guiding the Differentiation Direction of Pancreatic Islet-Derived Stem Cells by Glycated Collagen. Stem Cells Int 2018; 2018:6143081. [PMID: 30057625 PMCID: PMC6051021 DOI: 10.1155/2018/6143081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 11/17/2022] Open
Abstract
The microenvironment is an important factor of stem cells regulating their maintenance, survival, and differentiation. The glycation of proteins with reducing sugars through nonenzymatic reactions induces the collagen cross-linking, which causes tissue stiffening, which is enhanced during aging and diabetes. In this study, we aimed to analyze the effect of glycated collagen on the stem cell culture and differentiation. The collagen type 1 was modified by glycation with mannose, rhamnose, arabinose, and glucose. After the culture of mesenchymal stem cells on the coated surfaces with glycated collagen, the differences in cell adhesion, proliferation, and differentiation were compared. The results showed that the modifications did not induce apoptosis or cause cell death. However, the culture of cells on modified collagens improved the proliferation. It was found that the mannose-modified collagen stimulated the adipogenic differentiation of stem cells, and rhamnose-modified collagen supports the differentiation into both osteogenic and insulin-producing cells. The low concentration of monosaccharides during glycation process improved the characteristics of the matrix protein in favor of stem cell differentiation. Modification of the collagen by glycation might be used as a tool to improve natural polymers for material-induced stem cell differentiation in the future.
Collapse
Affiliation(s)
- Gokhan Duruksu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, 41380 Izmit, Kocaeli, Turkey
- Institute of Health Sciences, Stem Cell Department, Kocaeli University, 41380 Izmit, Kocaeli, Turkey
| | - Aysegul Aciksari
- Institute of Health Sciences, Stem Cell Department, Kocaeli University, 41380 Izmit, Kocaeli, Turkey
| |
Collapse
|
6
|
Liu Z, Speroni L, Quinn KP, Alonzo C, Pouli D, Zhang Y, Stuntz E, Sonnenschein C, Soto AM, Georgakoudi I. 3D organizational mapping of collagen fibers elucidates matrix remodeling in a hormone-sensitive 3D breast tissue model. Biomaterials 2018; 179:96-108. [PMID: 29980078 DOI: 10.1016/j.biomaterials.2018.06.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/08/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022]
Abstract
Hormones play an important role in normal and diseased breast tissue development. However, they can also disrupt cell-matrix interactions and their role in extracellular matrix reorganization during epithelial morphogenesis remains poorly understood, partly due to a lack of sensitive approaches for matrix characterization. Here, we assess the hormonal regulation of matrix reorganization in a three-dimensional (3D) breast tissue culture model using a novel metric, i.e., 3D directional variance, to characterize the 3D organization of collagen fibers visualized via high-resolution, second harmonic generation imaging. This metric enables resolving and quantifying patterns of spatial organization throughout the matrix surrounding epithelial structures treated with 17β-estradiol (E2) alone, and E2 in combination with either promegestone, a progestogen, or prolactin. Addition of promegestone results in the most disorganized fibers, while the E2 alone treatment leads to the most organized ones. Location-dependent organization mapping indicates that only the prolactin treatment leads to significant heterogeneities in the regional organization of collagen fibers, with higher levels of alignment observed at the end of the elongated epithelial structures. The observed collagen organization patterns for all groups persist for tens of micrometers. In addition, a comparison between 3D directional variance and typical 2D analysis approaches reveals an improved sensitivity of the 3D metric to identify organizational heterogeneities and differences among treatment groups. These results demonstrate that 3D directional variance is sensitive to subtle changes in the extracellular micro-environment and has the potential to elucidate reciprocal cell-matrix interactions in the context of numerous applications involving the study of normal and diseased tissue morphogenesis.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Speroni
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Carlo Alonzo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dimitra Pouli
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Emily Stuntz
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
7
|
Tajima S, Tabata Y. Preparation of epithelial cell aggregates incorporating matrigel microspheres to enhance proliferation and differentiation of epithelial cells. Regen Ther 2017; 7:34-44. [PMID: 30271850 PMCID: PMC6134895 DOI: 10.1016/j.reth.2017.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/10/2017] [Accepted: 07/04/2017] [Indexed: 11/25/2022] Open
Abstract
The objective of this study is to investigate the effect of matrigel microspheres (MM), gelatin hydrogel microspheres (GM), and matrigel-coated GM on the proliferated and biological functions of epithelial cells in cell aggregates incorporating the microspheres. The MM were prepared by a coacelvation method. When mammary epithelial EpH4 cells were cultured with the MM, GM, and matrigel-coated GM in round U-bottom wells of 96-multiwell culture plates which had been coated with poly (vinyl alcohol) (PVA) to suppress the cell adhesion, EpH4 cell aggregates with each microspheres homogeneously incorporated were formed. Higher EpH4 cells proliferation was observed for cell aggregates incorporating MM, GM, and matrigel-coated GM compared with the conventional 3-dimensional (3D) culture method. When examined to evaluate the epithelial differentiation of EpH4 cells, the β-casein expression was significantly higher for the cell aggregates incorporating MM than that of aggregates incorporating GM and matrigel-coated GM or the conventional 3D culture method. It is concluded that the proliferation and differentiation of mammary epithelial EpH4 cells were promoted by the incorporation of MM.
Collapse
Affiliation(s)
- Shuhei Tajima
- Department of Biomaterials, Institute for Life and Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Life and Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
8
|
Hillreiner M, Müller NI, Koch HM, Schmautz C, Küster B, Pfaffl MW, Kliem H. Establishment of a 3D cell culture model of primary bovine mammary epithelial cells extracted from fresh milk. In Vitro Cell Dev Biol Anim 2017. [PMID: 28643224 DOI: 10.1007/s11626-017-0169-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
For the investigation of molecular processes underlying diseases of the bovine mammary gland, primary bovine mammary epithelial cells (pbMEC) are used. They are known to contribute to the innate immune system of the bovine mammary gland. The functionality of pbMEC depends on the maintenance of in vivo characteristics. So far, the optimization of pbMEC culture conditions was intended in a variety of experiments. For this purpose, most of the studies used stable cell lines or primary cells obtained from udder biopsies of slaughtered animals. By contrast, within our study, pbMEC of healthy and first lactating Brown Swiss cows were non-invasively isolated from fresh milk. The non-invasively isolated pbMEC were cultivated on the extracellular matrix-like scaffold Matrigel®. Further, they were challenged with different compositions of proliferation media, containing lactogenic hormones and/or the essential amino acid L-lysine. Changes in expression levels of genes coding for milk proteins and for components of the janus kinase/signal transducers and activators of transcription (JAK-STAT) and mTOR pathways were analyzed by RT-qPCR. The secreted proteins were analyzed by LC-MS/MS measurements. We showed for the first time the establishment of a physiologically functional 3D cell culture model of pbMEC isolated from fresh milk. This represents a primary cell culture model system, based on non-invasive cell collection, that can be used to unravel physiological processes in an unbiased manner.
Collapse
Affiliation(s)
- Maria Hillreiner
- Chair of Animal Physiology and Immunology, Technische Universität München, Freising, Germany
| | - Nadine I Müller
- Chair of Animal Physiology and Immunology, Technische Universität München, Freising, Germany
| | - Heiner M Koch
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Christiane Schmautz
- Chair of Animal Physiology and Immunology, Technische Universität München, Freising, Germany
| | - Bernhard Küster
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany.,Bavarian Biomolecular Mass Spectrometry Center, Technische Universität München, Freising, Germany
| | - Michael W Pfaffl
- Chair of Animal Physiology and Immunology, Technische Universität München, Freising, Germany
| | - Heike Kliem
- Chair of Animal Physiology and Immunology, Technische Universität München, Freising, Germany.
| |
Collapse
|
9
|
Tajima S, Tabata Y. Preparation of EpH4 and 3T3L1 cells aggregates incorporating gelatin hydrogel microspheres for a cell condition improvement. Regen Ther 2017; 6:90-99. [PMID: 30271843 PMCID: PMC6134911 DOI: 10.1016/j.reth.2017.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
The objective of this study is to prepare three dimensional (3D) of mouse mammary epithelial EpH4 and mouse preadipocyte 3T3L1 cells in the presence of gelatin hydrogel microspheres (GM) and evaluate the effect of GM presence on the survival and functions of cells in the 3D cell aggregates. Gelatin was dehydrothermally crosslinked at 140 °C for 48 h in a water-in-oil emulsion state to obtain the GM with average diameters of 50 and 200 μm, followed by treatment with fibronectin (FN). EpH4 and/or 3T3L1 cells were cultured with or without the FN-treated GM in round U-bottom wells of 96-multiwell culture plates which had been coated with poly (vinyl alcohol) (PVA) to allow the cells to form their aggregates. On the other hand, EpH4 cells were precultured with the FN-treated GM, and then continued to culture with 3T3L1 cells in the same condition described above. The EpH4 cells attached onto the GM in the cell number dependent manner, irrespective of their size. When 3T3L1 cells were incubated with the original and GM-preincubated EpH4 cells in the presence of both the FN-treated GM, the number of alive cells in the aggregates was significantly high compared with that for the absence of FN-treated GM. In addition, higher β-casein expression level of EpH4 cells in EpH4/3T3L1 cells aggregates in the presence of FN-treated GM was observed than that of cells in the absence of FN-treated GM. Laminin secretion was also promoted for the cells aggregates cultured with FN-treated GM. It is concluded that the presence of FN-treated GM in the EpH4/3T3L1 cells aggregates gave a better condition to cells, resulting in an enhanced generation of β-casein from EpH4 cells in the aggregates.
Collapse
Affiliation(s)
- Shuhei Tajima
- Department of Biomaterials, Institute for Life and Frontier Medical Sciences, Kyoto University, 53 Kawara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Life and Frontier Medical Sciences, Kyoto University, 53 Kawara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
10
|
Shan L, Yu M, Snyderwine EG. Global Gene Expression Profiling of Chemically Induced Rat Mammary Gland Carcinomas and Adenomas. Toxicol Pathol 2016; 33:768-75. [PMID: 16316942 DOI: 10.1080/01926230500437027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chemical carcinogens induce both benign and malignant mammary gland tumors in female Sprague-Dawley rats. To identify gene expression profiles associated with malignancy, cDNA microarray analysis was used to compare gene expression profiles in rat mammary gland carcinomas, adenomas, and normal mammary gland. Tumors were induced with various chemical carcinogens including 2-amino-1-methyl-6-phenylimidazo[4,5- b]pyridine (PhIP), 2-amino-3,8-dimethylimidazo[4,5- f]quinoxaline (MeIQx), 7-12-dimethylbenz[ a]anthracene (DMBA), N-nitrosomethylurea (NMU), and 4-aminobiphenyl. The global gene expression profiles in carcinomas and adenomas were distinguishable by hierarchical clustering and multi-dimensional scaling analyses. Permutation analysis revealed 110 clones statistically differentially expressed between benign and malignant tumors ( p < 0.0005). Carcinomas showed relatively high expression of several genes associated with mammary epithelial cell growth and proliferation (e.g., cyclin D1, PDGFα) and relatively low expression of differentiation marker genes (e.g., β-casein, whey acidic protein, transferrin). Other categories of genes showing differential expression between carcinomas and adenomas were associated with protein homeostasis, cytoskeleton, extracellular matrix, and cell metabolism (fatty acid metabolism, oxidative phosphorylation, and glycolysis). Major gene families implicated in malignancy by over-expression in carcinomas included the annexins (annexin A1 and A4) and Stat family of transcription factors (Stat3 and Stat5a). The elevated expression of the prolactin receptor in carcinomas concomitant with several components of the mitogenic prolactin signaling pathway implicated prolactin/prolactin receptor/Stat5a/cyclin D1 in rat mammary gland malignancy.
Collapse
Affiliation(s)
- Liang Shan
- Chemical Carcinogenesis Section, Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892-4262, USA
| | | | | |
Collapse
|
11
|
Thorne JT, Segal TR, Chang S, Jorge S, Segars JH, Leppert PC. Dynamic reciprocity between cells and their microenvironment in reproduction. Biol Reprod 2014; 92:25. [PMID: 25411389 DOI: 10.1095/biolreprod.114.121368] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dynamic reciprocity (DR) refers to the ongoing, bidirectional interaction between cells and their microenvironment, specifically the extracellular matrix (ECM). The continuous remodeling of the ECM exerts mechanical force on cells and modifies biochemical mediators near the cell membrane, thereby initiating cell-signaling cascades that produce changes in gene expression and cell behavior. Cellular changes, in turn, affect the composition and organization of ECM components. These continuous interactions are the fundamental principle behind DR, and its critical role throughout development and adult tissue homeostasis has been extensively investigated. While DR in the mammary gland has been well described, we provide direct evidence that similar dynamic interactions occur in other areas of reproductive biology as well. In order to establish the importance of DR in the adaptive functioning of the female reproductive tract, we present our most current understanding of DR in reproductive tissues, exploring the mammary gland, ovary, and uterus. In addition to explaining normal physiological function, investigating DR may shed new light into pathologic processes that occur in these tissues and provide an exciting opportunity for novel therapeutic intervention.
Collapse
Affiliation(s)
- Jeffrey T Thorne
- Department of Obstetrics & Gynecology, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Thalia R Segal
- Department of Obstetrics & Gynecology, North Shore - Long Island Jewish Hospital, Manhasset, New York
| | - Sydney Chang
- Unit of Reproductive Endocrinology and Infertility, Eunice Kennedy Shriver National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland Department of Obstetrics & Gynecology, Duke University School of Medicine, Durham, North Carolina
| | - Soledad Jorge
- Unit of Reproductive Endocrinology and Infertility, Eunice Kennedy Shriver National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland Yale University School of Medicine, New Haven, Connecticut
| | - James H Segars
- Unit of Reproductive Endocrinology and Infertility, Eunice Kennedy Shriver National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland
| | - Phyllis C Leppert
- Department of Obstetrics & Gynecology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
12
|
Sonowal H, Kumar A, Bhattacharyya J, Gogoi PK, Jaganathan BG. Inhibition of actin polymerization decreases osteogeneic differentiation of mesenchymal stem cells through p38 MAPK pathway. J Biomed Sci 2013; 20:71. [PMID: 24070328 PMCID: PMC3849435 DOI: 10.1186/1423-0127-20-71] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/22/2013] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Mesenchymal Stem Cells (MSC) are important candidates for therapeutic applications due to their ex vivo proliferation and differentiation capacity. MSC differentiation is controlled by both intrinsic and extrinsic factors and actin cytoskeleton plays a major role in the event. In the current study, we tried to understand the initial molecular mechanisms and pathways that regulate the differentiation of MSC into osteocytes or adipocytes. RESULTS We observed that actin modification was important during differentiation and differentially regulated during adipogenesis and osteogenesis. Initial disruption of actin polymerization reduced further differentiation of MSC into osteocytes and osteogenic differentiation was accompanied by increase in ERK1/2 and p38 MAPK phosphorylation. However, only p38 MAPK phosphorylation was down regulated upon inhibition of actin polymerization which as accompanied by decreased CD49E expression. CONCLUSION Taken together, our results show that actin modification is a pre-requisite for MSC differentiation into osteocytes and adipocytes and osteogenic differentiation is regulated through p38 MAPK phosphorylation. Thus by modifying their cytoskeleton the differentiation potential of MSC could be controlled which might have important implications for tissue repair and regeneration.
Collapse
Affiliation(s)
- Himangshu Sonowal
- Stem Cell and Cancer Biology Group, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Atul Kumar
- Stem Cell and Cancer Biology Group, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Jina Bhattacharyya
- Department of Hematology, Gauhati Medical College Hospital, Guwahati, Assam, India
| | - Pabitra Kumar Gogoi
- Department of Hematology, Gauhati Medical College Hospital, Guwahati, Assam, India
| | - Bithiah Grace Jaganathan
- Stem Cell and Cancer Biology Group, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
13
|
Glukhova MA, Streuli CH. How integrins control breast biology. Curr Opin Cell Biol 2013; 25:633-41. [PMID: 23886475 PMCID: PMC3807876 DOI: 10.1016/j.ceb.2013.06.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/28/2013] [Accepted: 06/28/2013] [Indexed: 02/07/2023]
Abstract
This article explores new ideas about how the ECM-integrin axis controls normal and malignant breast biology. We discuss the role of integrins in mammary stem cells, and how cell-matrix interactions regulate ductal and alveolar development and function. We also examine the contribution of integrins to tissue disorganisation and metastasis, and how an altered stromal and ECM tumour microenvironment affects the cancer cell niche both within primary tumours and at distant sites. Finally, we mention novel strategies for integrin-directed breast cancer treatment.
Collapse
Affiliation(s)
- Marina A Glukhova
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | | |
Collapse
|
14
|
Tamarit B, Bugault F, Pillet AH, Lavergne V, Bochet P, Garin N, Schwarz U, Thèze J, Rose T. Membrane microdomains and cytoskeleton organization shape and regulate the IL-7 receptor signalosome in human CD4 T-cells. J Biol Chem 2013; 288:8691-8701. [PMID: 23329834 DOI: 10.1074/jbc.m113.449918] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)-7 is the main homeostatic regulator of CD4 T-lymphocytes (helper) at both central and peripheral levels. Upon activation by IL-7, several signaling pathways, mainly JAK/STAT, PI3K/Akt and MAPK, induce the expression of genes involved in T-cell differentiation, activation, and proliferation. We have analyzed the early events of CD4 T-cell activation by IL-7. We have shown that IL-7 in the first few min induces the formation of cholesterol-enriched membrane microdomains that compartmentalize its activated receptor and initiate its anchoring to the cytoskeleton, supporting the formation of the signaling complex, the signalosome, on the IL-7 receptor cytoplasmic domains. Here we describe by stimulated emission depletion microscopy the key roles played by membrane microdomains and cytoskeleton transient organization in the IL-7-regulated JAK/STAT signaling pathway. We image phospho-STAT5 and cytoskeleton components along IL-7 activation kinetics using appropriate inhibitors. We show that lipid raft inhibitors delay and reduce IL-7-induced JAK1 and JAK3 phosphorylation. Drug-induced disassembly of the cytoskeleton inhibits phospho-STAT5 formation, transport, and translocation into the nucleus that controls the transcription of genes involved in T-cell activation and proliferation. We fit together the results of these quantitative analyses and propose the following mechanism. Activated IL-7 receptors embedded in membrane microdomains induce actin-microfilament meshwork formation, anchoring microtubules that grow radially from rafted receptors to the nuclear membrane. STAT5 phosphorylated by signalosomes are loaded on kinesins and glide along the microtubules across the cytoplasm to reach the nucleus 2 min after IL-7 stimulation. Radial microtubules disappear 15 min later, while transversal microtubules, independent of phospho-STAT5 transport, begin to bud from the microtubule organization center.
Collapse
Affiliation(s)
- Blanche Tamarit
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France; Université Pierre et Marie Curie, Cellule Pasteur-UPMC, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Florence Bugault
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Anne-Hélène Pillet
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France; Université Pierre et Marie Curie, Cellule Pasteur-UPMC, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Vincent Lavergne
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Pascal Bochet
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR3525, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Nathalie Garin
- Leica Microsystems AG, Max Schmidheiny Strasse 201, CH-9435 Heerbrugg, Switzerland
| | - Ulf Schwarz
- Leica Microsystems CMS GmbH, Am Friedrichplatz, D-68165 Mannheim, Germany
| | - Jacques Thèze
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Thierry Rose
- Institut Pasteur, Département Infection et Epidémiologie, Département d'Immunologie, Unité d'Immunogénétique Cellulaire, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
15
|
Stromal-epithelial interactions modulate cross-talk between prolactin receptor and HER2/Neu in breast cancer. Breast Cancer Res Treat 2012; 134:157-69. [PMID: 22270933 DOI: 10.1007/s10549-012-1954-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 01/06/2012] [Indexed: 12/14/2022]
Abstract
Prolactin (PRL) promotes the proliferation and survival of breast cancer cells in part via the transactivation of human epidermal growth factor receptor 2 (HER2), also known as Neu in rodents. A PRL receptor (PRLR) antagonist, G129R, has been developed, which indirectly inhibits the tyrosine phosphorylation of HER2 (p-HER2) in human breast cancer cell lines. In this study, we investigate the effects of cancer-associated fibroblasts (CAFs) upon this molecular cross-talk using tumor cells and CAFs derived from spontaneous mammary tumors of female MMTV-neu transgenic mice. Tumors were resected and cultured as small tumor chunks (~3 mm3) or were cultured in monolayer. G129R reduced tyrosine phosphorylation of Neu (p-Neu) in a dose-dependent manner (IC50~10 μg/ml) in tumor chunks, but had no effect on primary tumor epithelial cells grown in monolayer. Direct co-culture of mouse or human tumor epithelial cell lines with CAFs restored the epithelial cells' response to G129R, similar to that observed in mouse tumor chunks. The addition of PRL, as expected, induced p-Neu in both the tumor chunk and co-culture models. The inhibitory effect of G129R was absent when CAFs were physically separated from mouse tumor epithelial cells using a transwell system, or when CAFs were replaced with normal fibroblasts in direct co-culture with human or mouse tumor epithelial cells. In vivo, G129R reduced p-Neu levels in primary mammary tumors of mice in a time- and dose-dependent manner. In conclusion, CAFs play a critical role in bridging the cross-talk between PRL and HER2/Neu in both mouse and human models of breast cancer. The inhibitory effects of G129R on p-Neu and on tumor growth are dependent upon interactions of tumor epithelial cells with CAFs.
Collapse
|
16
|
Talhouk RS, Khalil AA, Bajjani R, Rahme GJ, El-Sabban ME. Gap junctions mediate STAT5-independent β-casein expression in CID-9 mammary epithelial cells. ACTA ACUST UNITED AC 2011; 18:104-16. [DOI: 10.3109/15419061.2011.639468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Rabih S. Talhouk
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Antoine A. Khalil
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Rachid Bajjani
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Gilbert J. Rahme
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Marwan E. El-Sabban
- Department of Human Morphology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
17
|
Weymouth N, Shi Z, Rockey DC. Smooth muscle α actin is specifically required for the maintenance of lactation. Dev Biol 2011; 363:1-14. [PMID: 22123032 DOI: 10.1016/j.ydbio.2011.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 11/30/2022]
Abstract
Smooth muscle α-actin (Acta2) is one of six highly conserved mammalian actin isoforms that appear to exhibit functional redundancy. Nonetheless, we have postulated a specific functional role for the smooth muscle specific isoform. Here, we show that Acta2 deficient mice have a remarkable mammary phenotype such that dams lacking Acta2 are unable to nurse their offspring effectively. The phenotype was rescued in cross fostering experiments with wild type mice, excluding a developmental defect in Acta2 null pups. The mechanism for the underlying phenotype is due to myoepithelial dysfunction postpartum resulting in precocious involution. Further, we demonstrate a specific defect in myoepithelial cell contractility in Acta2 null mammary glands, despite normal expression of cytoplasmic actins. We conclude that Acta2 specifically mediates myoepithelial cell contraction during lactation and that this actin isoform therefore exhibits functional specificity.
Collapse
Affiliation(s)
- Nate Weymouth
- University of Texas Southwestern Medical Center, Dallas, TX 75390-8887, USA
| | | | | |
Collapse
|
18
|
Spencer VA, Costes S, Inman JL, Xu R, Chen J, Hendzel MJ, Bissell MJ. Depletion of nuclear actin is a key mediator of quiescence in epithelial cells. J Cell Sci 2011; 124:123-32. [PMID: 21172822 DOI: 10.1242/jcs.073197] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Functional differentiation is orchestrated by precise growth-regulatory controls conveyed by the tissue microenvironment. Cues from laminin 111 (LN1) lower transcription and suppress mammary epithelial cell growth in culture, but how LN1 induces quiescence is unknown. Recent literature points to involvement of nuclear β-actin in transcriptional regulation. Here, we show that quiescence induced by growth factor withdrawal, or LN1 addition, rapidly decreases nuclear β-actin. LN1, but not other extracellular matrix (ECM) molecules, decreases the levels of nuclear β-actin and destabilizes RNA polymerase (RNA Pol) II and III binding to transcription sites, leading to a dramatic drop in transcription and DNA synthesis. Constitutive overexpression of globular β-actin in the nucleus reverses the effect of LN1 on transcription and RNA Pol II association and prevents the cells from becoming quiescent in the presence of LN1. The physiological relevance of our findings was verified by identifying a clear spatial separation of LN1 and β-actin in developing mammary end buds. These data indicate a novel role for nuclear β-actin in growth arrest of epithelial cells and underscore the importance of the integrity of the basement membrane in homeostasis.
Collapse
Affiliation(s)
- Virginia A Spencer
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 977R225A, Berkeley, CA 94720, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Storch K, Eke I, Borgmann K, Krause M, Richter C, Becker K, Schröck E, Cordes N. Three-dimensional cell growth confers radioresistance by chromatin density modification. Cancer Res 2010; 70:3925-34. [PMID: 20442295 DOI: 10.1158/0008-5472.can-09-3848] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cell shape and architecture are determined by cell-extracellular matrix interactions and have profound effects on cellular behavior, chromatin condensation, and tumor cell resistance to radiotherapy and chemotherapy. To evaluate the role of chromatin condensation for radiation cell survival, tumor cells grown in three-dimensional (3D) cell cultures as xenografts and monolayer cell cultures were compared. Here, we show that increased levels of heterochromatin in 3D cell cultures characterized by histone H3 deacetylation and induced heterochromatin protein 1alpha expression result in increased radiation survival and reduced numbers of DNA double strand breaks (DSB) and lethal chromosome aberrations. Intriguingly, euchromatin to heterochromatin-associated DSBs were equally distributed in irradiated 3D cell cultures and xenograft tumors, whereas irradiated monolayer cultures showed a 2:1 euchromatin to heterochromatin DSB distribution. Depletion of histone deacetylase (HDAC) 1/2/4 or application of the class I/II pharmacologic HDAC inhibitor LBH589 induced moderate or strong chromatin decondensation, respectively, which was translated into cell line-dependent radiosensitization and, in case of LBH589, into an increased number of DSBs. Neither growth conditions nor HDAC modifications significantly affected the radiation-induced phosphorylation of the important DNA repair protein ataxia telangiectasia mutated. Our data show an interrelation between cell morphology and cellular radiosensitivity essentially based on chromatin organization. Understanding the molecular mechanisms by which chromatin structure influences the processing of radiation-induced DNA lesions is of high relevance for normal tissue protection and optimization of cancer therapy.
Collapse
Affiliation(s)
- Katja Storch
- OncoRay-Center for Radiation Research in Oncology, Department of Radiation Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Spencer VA, Xu R, Bissell MJ. Gene expression in the third dimension: the ECM-nucleus connection. J Mammary Gland Biol Neoplasia 2010; 15:65-71. [PMID: 20107877 PMCID: PMC2912292 DOI: 10.1007/s10911-010-9163-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 01/05/2010] [Indexed: 12/25/2022] Open
Abstract
Decades ago, we and others proposed that the dynamic interplay between a cell and its surrounding environment dictates cell phenotype and tissue structure. Whereas much has been discovered about the effects of extracellular matrix molecules on cell growth and tissue-specific gene expression, the nuclear mechanisms through which these molecules promote these physiological events remain unknown. Using mammary epithelial cells as a model, the purpose of this review is to discuss how the extracellular matrix influences nuclear structure and function in a three-dimensional context to promote epithelial morphogenesis and function in the mammary gland.
Collapse
|
21
|
Rose T, Pillet AH, Lavergne V, Tamarit B, Lenormand P, Rousselle JC, Namane A, Thèze J. Interleukin-7 compartmentalizes its receptor signaling complex to initiate CD4 T lymphocyte response. J Biol Chem 2010; 285:14898-14908. [PMID: 20167604 DOI: 10.1074/jbc.m110.104232] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL)-7 is a central cytokine that controls homeostasis of the CD4 T lymphocyte pool. Here we show on human primary cells that IL-7 binds to preassembled receptors made up of proprietary chain IL-7Ralpha and the common chain gammac shared with IL-2, -4, -9, -15, and -21 receptors. Upon IL-7 binding, both chains are driven in cholesterol- and sphingomyelin-rich rafts where associated signaling proteins Jak1, Jak3, STAT1, -3, and -5 are found to be phosphorylated. Meanwhile the IL-7.IL-7R complex interacts with the cytoskeleton that halts its diffusion as measured by single molecule fluorescence autocorrelated spectroscopy monitored by microimaging. Comparative immunoprecipitations of IL-7Ralpha signaling complex from non-stimulated and IL-7-stimulated cells confirmed recruitment of proteins such as STATs, but many others were also identified by mass spectrometry from two-dimensional gels. Among recruited proteins, two-thirds are involved in cytoskeleton and raft formation. Thus, early events leading to IL-7 signal transduction involve its receptor compartmentalization into membrane nanodomains and cytoskeleton recruitment.
Collapse
Affiliation(s)
- Thierry Rose
- Institut Pasteur, Unité d'Immunogénétique Cellulaire, Département Infection et Epidémiologie, Département d'Immunologie, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France.
| | - Anne-Hélène Pillet
- Institut Pasteur, Unité d'Immunogénétique Cellulaire, Département Infection et Epidémiologie, Département d'Immunologie, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Vincent Lavergne
- Institut Pasteur, Unité d'Immunogénétique Cellulaire, Département Infection et Epidémiologie, Département d'Immunologie, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Blanche Tamarit
- Institut Pasteur, Unité d'Immunogénétique Cellulaire, Département Infection et Epidémiologie, Département d'Immunologie, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Pascal Lenormand
- Plate-Forme Protéomique, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | | | - Abdelkader Namane
- Plate-Forme Protéomique, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Jacques Thèze
- Institut Pasteur, Unité d'Immunogénétique Cellulaire, Département Infection et Epidémiologie, Département d'Immunologie, Genopole, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
22
|
Spencer VA, Xu R, Bissell MJ. Extracellular matrix, nuclear and chromatin structure, and gene expression in normal tissues and malignant tumors: a work in progress. Adv Cancer Res 2009; 97:275-94. [PMID: 17419950 PMCID: PMC2912285 DOI: 10.1016/s0065-230x(06)97012-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Almost three decades ago, we presented a model where the extracellular matrix (ECM) was postulated to influence gene expression and tissue-specificity through the action of ECM receptors and the cytoskeleton. This hypothesis implied that ECM molecules could signal to the nucleus and that the unit of function in higher organisms was not the cell alone, but the cell plus its microenvironment. We now know that ECM invokes changes in tissue and organ architecture and that tissue, cell, nuclear, and chromatin structure are changed profoundly as a result of and during malignant progression. Whereas some evidence has been generated for a link between ECM-induced alterations in tissue architecture and changes in both nuclear and chromatin organization, the manner by which these changes actively induce or repress gene expression in normal and malignant cells is a topic in need of further attention. Here, we will discuss some key findings that may provide insights into mechanisms through which ECM could influence gene transcription and how tumor cells acquire the ability to overcome these levels of control.
Collapse
Affiliation(s)
- Virginia A Spencer
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | | | |
Collapse
|
23
|
Xu R, Boudreau A, Bissell MJ. Tissue architecture and function: dynamic reciprocity via extra- and intra-cellular matrices. Cancer Metastasis Rev 2009; 28:167-76. [PMID: 19160017 DOI: 10.1007/s10555-008-9178-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mammary gland development, functional differentiation, and homeostasis are orchestrated and sustained by a balance of biochemical and biophysical cues from the organ's microenvironment. The three-dimensional microenvironment of the mammary gland, predominantly 'encoded' by a collaboration between the extracellular matrix (ECM), hormones, and growth factors, sends signals from ECM receptors through the cytoskeletal intracellular matrix to nuclear and chromatin structures resulting in gene expression; the ECM in turn is regulated and remodeled by signals from the nucleus. In this chapter, we discuss how coordinated ECM deposition and remodeling is necessary for mammary gland development, how the ECM provides structural and biochemical cues necessary for tissue-specific function, and the role of the cytoskeleton in mediating the extra--to intracellular dialogue occurring between the nucleus and the microenvironment. When operating normally, the cytoskeletal-mediated dynamic and reciprocal integration of tissue architecture and function directs mammary gland development, tissue polarity, and ultimately, tissue-specific gene expression. Cancer occurs when these dynamic interactions go awry for an extended time.
Collapse
Affiliation(s)
- Ren Xu
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 977-225A, Berkeley, CA 94720, USA.
| | | | | |
Collapse
|
24
|
Abstract
The Wiskott-Aldrich syndrome (WAS) interacting protein (WIP) stabilizes the WAS protein (WASP), the product of the gene mutated in WAS. WIP-deficient T cells have low WASP levels, limiting the usefulness of WIP KO mice in defining the role of WIP in T cell function. To define this role, we compared WIP/WASP double KO (DKO) mice to WASP KO mice on DO11.10 background. T cell development was normal in both strains, but peripheral T cell numbers were significantly decreased in DKO mice. WASP KO T cells proliferated and secreted IL-2 normally in response to OVA peptide (OVAp). In contrast, T cells from DKO mice proliferated poorly in response to OVAp in vitro, and cutaneous hapten hypersensitivity was deficient in these mice. DKO T cells up-regulated CD25 expression and secreted normal amounts of IL-2 after antigen stimulation, but had defective response to IL-2, evidenced by failure to further up-regulate CD25 expression, phosphorylate STAT5, and induce expression of STAT5-dependent genes. DKO, but not WASP KO, T cells had a disrupted subcortical actin cytoskeleton and impaired actin polymerization after T cell antigen receptor (TCR) ligation. These results indicate that WIP is essential for IL-2 signaling and responsiveness in T cells, possibly because of its critical role in TCR-triggered actin cytoskeletal reorganization.
Collapse
|
25
|
Akhtar N, Marlow R, Lambert E, Schatzmann F, Lowe ET, Cheung J, Katz E, Li W, Wu C, Dedhar S, Naylor MJ, Streuli CH. Molecular dissection of integrin signalling proteins in the control of mammary epithelial development and differentiation. Development 2009; 136:1019-27. [PMID: 19211680 DOI: 10.1242/dev.028423] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cell-matrix adhesion is essential for the development and tissue-specific functions of epithelia. For example, in the mammary gland, beta1-integrin is necessary for the normal development of alveoli and for the activation of endocrine signalling pathways that determine cellular differentiation. However, the adhesion complex proteins linking integrins with downstream effectors of hormonal signalling pathways are not known. To understand the mechanisms involved in connecting adhesion with this aspect of cell phenotype, we examined the involvement of two proximal beta1-integrin signalling intermediates, integrin-linked kinase (ILK) and focal adhesion kinase (FAK). By employing genetic analysis using the Cre-LoxP system, we provide evidence that ILK, but not FAK, has a key role in lactogenesis in vivo and in the differentiation of cultured luminal epithelial cells. Conditional deletion of ILK both in vivo and in primary cell cultures resulted in defective differentiation, by preventing phosphorylation and nuclear translocation of STAT5, a transcription factor required for lactation. Expression of an activated RAC (RAS-related C3 botulinum substrate) in ILK-null acini restored the lactation defect, indicating that RAC1 provides a mechanistic link between the integrin/ILK adhesion complex and the differentiation pathway. Thus, we have determined that ILK is an essential downstream component of integrin signalling involved in differentiation, and have identified a high degree of specificity within the integrin-based adhesome that links cell-matrix interactions with the tissue-specific function of epithelia.
Collapse
Affiliation(s)
- Nasreen Akhtar
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Alcaraz J, Xu R, Mori H, Nelson CM, Mroue R, Spencer VA, Brownfield D, Radisky DC, Bustamante C, Bissell MJ. Laminin and biomimetic extracellular elasticity enhance functional differentiation in mammary epithelia. EMBO J 2008; 27:2829-38. [PMID: 18843297 PMCID: PMC2569873 DOI: 10.1038/emboj.2008.206] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 09/15/2008] [Indexed: 01/08/2023] Open
Abstract
In the mammary gland, epithelial cells are embedded in a ‘soft' environment and become functionally differentiated in culture when exposed to a laminin-rich extracellular matrix gel. Here, we define the processes by which mammary epithelial cells integrate biochemical and mechanical extracellular cues to maintain their differentiated phenotype. We used single cells cultured on top of gels in conditions permissive for β-casein expression using atomic force microscopy to measure the elasticity of the cells and their underlying substrata. We found that maintenance of β-casein expression required both laminin signalling and a ‘soft' extracellular matrix, as is the case in normal tissues in vivo, and biomimetic intracellular elasticity, as is the case in primary mammary epithelial organoids. Conversely, two hallmarks of breast cancer development, stiffening of the extracellular matrix and loss of laminin signalling, led to the loss of β-casein expression and non-biomimetic intracellular elasticity. Our data indicate that tissue-specific gene expression is controlled by both the tissues' unique biochemical milieu and mechanical properties, processes involved in maintenance of tissue integrity and protection against tumorigenesis.
Collapse
Affiliation(s)
- Jordi Alcaraz
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Stiening C, Hoying J, Abdallah M, Hoying A, Pandey R, Greer K, Collier R. The Effects of Endocrine and Mechanical Stimulation on Stage I Lactogenesis in Bovine Mammary Epithelial Cells. J Dairy Sci 2008; 91:1053-66. [DOI: 10.3168/jds.2007-0161] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
28
|
Bratthauer GL, Strauss BL, Barner R. Reversed Expression of the JAK/STAT Pathway Related Proteins Prolactin Receptor and STAT5a in Normal and Abnormal Breast Epithelial Cells. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2008; 1:7-14. [PMID: 21655368 PMCID: PMC3091403 DOI: 10.4137/bcbcr.s549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The JAK/STAT pathway is important for cellular metabolism. One component, STAT5a, is activated in the breast upon prolactin to prolactin receptor (PRLR) binding facilitating the transcription of genes involved in lobule development. STAT5a was previously found to be expressed in most normal breast epithelial cells but not in many in situ or invasive carcinomas except secretory carcinomas which retain STAT5a expression. This report examines the JAK/STAT pathway in the breast through the detection of PRLR and STAT5a. Fifty breast tissues, including benign secretory change, microglandular adenosis, usual and atypical hyperplasia and in situ and invasive ductal carcinoma both usual and secretory, were obtained from the files of the Armed Forces Institute of Pathology. Sections were immunostained with antibodies to PRLR and STAT5a. PRLR was minimally detected on the surface of a few normal breast epithelial cells whereas STAT5a was greatly expressed in over 80% of normal cell nuclei. PRLR was also minimally detected in secretory carcinomas expressing STAT5a. However, the opposite pattern was seen in breast carcinomas lacking STAT5a expression. PRLR was abundantly expressed in these cells. This reversed expression may indicate a JAK/STAT pathway disturbance that could play a role in the initiation or maintenance of an abnormal breast phenotype.
Collapse
Affiliation(s)
- Gary L Bratthauer
- Department of Gynecologic and Breast Pathology, Armed Forces Institute of Pathology, Washington
| | | | | |
Collapse
|
29
|
Watkin H, Richert MM, Lewis A, Terrell K, McManaman JP, Anderson SM. Lactation failure in Src knockout mice is due to impaired secretory activation. BMC DEVELOPMENTAL BIOLOGY 2008; 8:6. [PMID: 18215306 PMCID: PMC2266720 DOI: 10.1186/1471-213x-8-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 01/23/2008] [Indexed: 11/17/2022]
Abstract
Background Mammary gland development culminates in lactation and is orchestrated by numerous stimuli and signaling pathways. The Src family of nonreceptor tyrosine kinases plays a pivotal role in cell signaling. In order to determine if Src plays a role in mammary gland development we have examined mammary gland development and function during pregnancy and lactation in mice in which expression of Src has been eliminated. Results We have characterized a lactation defect in the Src-/- mice which results in the death of over 80% of the litters nursed by Src-/- dams. Mammary gland development during pregnancy appears normal in these mice; however secretory activation does not seem to occur. Serum prolactin levels are normal in Src-/- mice compared to wildtype controls. Expression of the prolactin receptor at both the RNA and protein level was decreased in Src-/- mice following the transition from pregnancy to lactation, as was phosphorylation of STAT5 and expression of milk protein genes. These results suggest that secretory activation, which occurs following parturition, does not occur completely in Src-/- mice. Failed secretory activation results in precocious involution in the mammary glands of Src-/- even when pups were suckling. Involution was accelerated following pup withdrawal perhaps as a result of incomplete secretory activation. In vitro differentiation of mammary epithelial cells from Src-/- mice resulted in diminished production of milk proteins compared to the amount of milk proteins produced by Src+/+ cells, indicating a direct role for Src in regulating the transcription/translation of milk protein genes in mammary epithelial cells. Conclusion Src is an essential signaling modulator in mammary gland development as Src-/- mice exhibit a block in secretory activation that results in lactation failure and precocious involution. Src appears to be required for increased expression of the prolactin receptor and successful downstream signaling, and alveolar cell organization.
Collapse
Affiliation(s)
- Harriet Watkin
- Department of Pathology, University of Colorado Health Sciences Center, Research Complex I, South Tower, Mail Stop 8104, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Ernens I, Clegg R, Schneider YJ, Larondelle Y. Short communication: Ability of cultured mammary epithelial cells in a bicameral system to secrete milk fat. J Dairy Sci 2007; 90:677-81. [PMID: 17235143 DOI: 10.3168/jds.s0022-0302(07)71550-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mammary epithelial cells from lactating cows were cultured onto inserts coated with type I collagen. Every second day, the rates of fatty acid synthesis and secretion were determined by measuring the amount of [14C]-labeled sodium acetate incorporated into lipids over a 4-h period. The [14C]-containing lipids were identified by thin layer chromatography fractionation. In parallel, the integrity of the cell layer was evaluated by measurement of transepithelial electrical resistance. The integrity increased progressively to reach a maximum after 8 d of culture. Cells incorporated acetate into lipids; 1.34% of acetate was incorporated into lipids produced by freshly isolated cells. This percentage decreased to 0.5% after 2 d of culture. Moreover, this capacity decreased with the duration in culture; on d 8, the rate of incorporation dropped to about 3% of that on d 2. In the cell extracts, the [14C]-labeled lipids were mainly triglycerides, although the proportion of diglycerides and phospholipids progressively increased as a part of total newly synthesized lipids. The proportion of triglycerides decreased 0.66 times between d 2 and 8 when the proportion of diglycerides and phospholipids increased 1.33 and 2.18 times, respectively. About 28% of the newly synthesized lipids were secreted within 4 h of incubation. Around 65 to 85% of these labeled lipids were found in the apical compartment, suggesting a partially vectorial secretion. But 58 to 80% of labeled lipids found in the apical and basolateral medium were free fatty acids. Functional tight junctions and incorporation of labeled fatty acids into triglycerides are not compatible with an inferred status of complete dedifferentiation of the cell layer. Moreover, triglyceride secretion seems compromised, probably due to the lack of an appropriate cell environment and cell shape.
Collapse
Affiliation(s)
- I Ernens
- Hannah Research Institute, Hannah Research Park, Ayr, KA6 5HL, Scotland, UK.
| | | | | | | |
Collapse
|
31
|
Akhtar N, Streuli CH. Rac1 links integrin-mediated adhesion to the control of lactational differentiation in mammary epithelia. ACTA ACUST UNITED AC 2006; 173:781-93. [PMID: 16754961 PMCID: PMC2063893 DOI: 10.1083/jcb.200601059] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The expression of tissue-specific genes during mammary gland differentiation relies on the coincidence of two distinct signaling events: the continued engagement of β1 integrins with the extracellular matrix (ECM) and a hormonal stimulus from prolactin (Prl). How the integrin and Prl receptor (PrlR) systems integrate to regulate milk protein gene synthesis is unknown. In this study, we identify Rac1 as a key link. Dominant-negative Rac1 prevents Prl-induced synthesis of the milk protein β-casein in primary mammary epithelial cells cultured as three-dimensional acini on basement membrane. Conversely, activated Rac1 rescues the defective β-casein synthesis that occurs under conditions not normally permissive for mammary differentiation, either in β1 integrin–null cells or in wild-type cells cultured on collagen. Rac1 is required downstream of integrins for activation of the PrlR/Stat5 signaling cascade. Cdc42 is also necessary for milk protein synthesis but functions via a distinct mechanism to Rac1. This study identifies the integration of signals provided by ECM and hormones as a novel role for Rho family guanosine triphosphatases.
Collapse
Affiliation(s)
- Nasreen Akhtar
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, England, United Kingdom
| | | |
Collapse
|
32
|
Faraci-Orf E, McFadden C, Vogel WF. DDR1 signaling is essential to sustain Stat5 function during lactogenesis. J Cell Biochem 2006; 97:109-21. [PMID: 16167341 DOI: 10.1002/jcb.20618] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Postnatal development of the mammary gland is achieved by an interplay of endocrine and extracellular matrix-derived signals. Despite intense research, a comprehensive understanding of the temporal and spatial coordination of these hormonal and basement membrane stimuli is still lacking. Here, we address the role of the collagen-receptor DDR1 in integrating extracellular matrix-derived signaling with the lactogenic pathway initiated by the prolactin receptor. We found that stimulation of DDR1-overexpressing mammary epithelial HC11 cells with collagen and prolactin resulted in stronger and more sustained induction of Stat5 phosphorylation as compared to control cells. Enhanced Stat5 activity in HC11-DDR1 cells correlated with increased beta-casein gene expression. In contrast, cells derived from DDR1-null mice showed reduced Stat5 activation upon lactogenic stimulation and completely failed to induce beta-casein expression. The cell-autonomous role of DDR1 in controlling ductal branching and alveologenesis prior to the onset of lactogenesis was corroborated by mammary tissue transplantation experiments. Our results show that aside from hormone- and cytokine receptors, DDR1 signaling establishes a third matrix-derived pathway vital to maintain mammary gland function.
Collapse
Affiliation(s)
- Elena Faraci-Orf
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | |
Collapse
|
33
|
Lopez-Perez M, Salazar EP. A role for the cytoskeleton in STAT5 activation in MCF7 human breast cancer cells stimulated with EGF. Int J Biochem Cell Biol 2006; 38:1716-28. [PMID: 16765629 DOI: 10.1016/j.biocel.2006.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 03/17/2006] [Accepted: 04/02/2006] [Indexed: 11/25/2022]
Abstract
A rapid increase in the tyrosine phosphorylation of signal transducers and activators of transcription (STAT) proteins has been extensively documented in cells stimulated with cytokines and growth factors. However, the mechanisms by which these transcription factors translocate to the nucleus have not been studied in detail. Our results demonstrate that stimulation of MCF7 cells with epidermal growth factor (EGF) promoted an increase in the phosphorylation of STAT5 at Tyr-694, as revealed by site-specific antibodies that recognized the phosphorylated state of this residue. In addition, EGF stimulated STAT5 nuclear translocation and an increased in STAT5 DNA binding activity. Prevention of microtubules and microfilaments polymerization induced a partial inhibition of STAT5 nuclear translocation and STAT5 DNA binding activity. However, STAT5 phosphorylation at Tyr-694 was dependent on the integrity of microtubule network and it was independent of the integrity of actin cytoskeleton. Furthermore, EGF induced the formation of the associations STAT5-tubulin and STAT5-kinesin heavy chain in a fashion dependent of cytoskeleton integrity. In summary, our results demonstrate, for the first time, that cytoskeleton plays an important role in STAT5 activation and translocation into the nucleus in MCF7 cells stimulated with EGF.
Collapse
Affiliation(s)
- Mario Lopez-Perez
- Departamento de Biologia Celular, Cinvestav-IPN, Av IPN# 2508, San Pedro Zacatenco, Mexico, DF 07360, Mexico
| | | |
Collapse
|
34
|
Holland MS, Holland RE. The cellular perspective on mammary gland development: stem/progenitor cells and beyond. J Dairy Sci 2006; 88 Suppl 1:E1-8. [PMID: 15876573 DOI: 10.3168/jds.s0022-0302(05)73132-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Study of the mammary gland at the stem cell level is necessary for understanding mammary gland development. Knowledge of mammary gland development and growth is the first step toward formulating strategies to improve milk production. The success of these strategies requires an understanding of the dynamics of adult stem cells and their progeny in the development of the bovine mammary gland. The stem cell lineage pathway begins with adult stem cells and ends with the production of terminally differentiated cells. The progression of adult stem cells along the mammary gland stem cell lineage pathway requires the coordination of many events. One important event in this process is cell differentiation. This differentiation process evolves with a gradient appearance of cell organelles progressing from stem cells to terminally differentiated cells. To dissect differentiation, mechanisms that regulate stem cells to differentiate toward a particular cell fate must be identified. Ultrastructural characteristics assist in distinguishing cells in various stages of differentiation in the mammary gland cell lineage pathway. Cells in the lineage pathway can become either epithelial cells or myoepithelial cells. Epithelial cells function in the production and secretion of milk, whereas myoepithelial cells assist epithelial cells in milk secretion. This review focuses on current concepts regarding adult stem cells and the recent progress on bovine mammary gland stem/progenitor cell development and differentiation. Multistep strategies that incorporate manipulation of the mechanisms influencing lineage choices in the mammary gland will produce beneficial effects on mammary gland development and milk production.
Collapse
Affiliation(s)
- M S Holland
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, 50011, USA.
| | | |
Collapse
|
35
|
Nelson CM, Bissell MJ. Modeling dynamic reciprocity: engineering three-dimensional culture models of breast architecture, function, and neoplastic transformation. Semin Cancer Biol 2005; 15:342-52. [PMID: 15963732 PMCID: PMC2933210 DOI: 10.1016/j.semcancer.2005.05.001] [Citation(s) in RCA: 209] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to understand why cancer develops as well as predict the outcome of pharmacological treatments, we need to model the structure and function of organs in culture so that our experimental manipulations occur under physiological contexts. This review traces the history of the development of a prototypic example, the three-dimensional (3D) model of the mammary gland acinus. We briefly describe the considerable information available on both normal mammary gland function and breast cancer generated by the current model and present future challenges that will require an increase in its complexity. We propose the need for engineered tissues that faithfully recapitulate their native structures to allow a greater understanding of tissue function, dysfunction, and potential therapeutic intervention.
Collapse
Affiliation(s)
| | - Mina J. Bissell
- Corresponding author. Tel.: +1 510 486 4365; fax: +1 510 486 5586. (M.J. Bissell)
| |
Collapse
|
36
|
Hinoue A, Takigawa T, Miura T, Nishimura Y, Suzuki S, Shiota K. Disruption of actin cytoskeleton and anchorage-dependent cell spreading induces apoptotic death of mouse neural crest cells cultured in vitro. ACTA ACUST UNITED AC 2005; 282:130-7. [PMID: 15627983 DOI: 10.1002/ar.a.20150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In vertebrate embryos, neural crest cells emigrate out of the neural tube and contribute to the formation of a variety of neural and nonneural tissues. Some neural crest cells undergo apoptotic death during migration, but its biological significance and the underlying mechanism are not well understood. We carried out an in vitro study to examine how the morphology and survival of cranial neural crest (CNC) cells of the mouse embryo are affected when their actin cytoskeleton or anchorage-dependent cell spreading is perturbed. Disruption of actin fiber organization by cytochalasin D (1 microg/ml) and inhibition of cell attachment by matrix metalloproteinase-2 (MMP-2; 2.0 units/ml) were followed by morphologic changes and apoptotic death of cultured CNC cells. When the actin cytoskeleton was disrupted by cytochalasin D, the morphologic changes of cultured CNC cells preceded DNA fragmentation. These results indicate that the maintenance of cytoskeleton and anchorage-dependent cell spreading are required for survival of CNC cells. The spatially and temporally regulated expression of proteinases may be essential for the differentiation and migration of neural crest cells.
Collapse
Affiliation(s)
- Atsushi Hinoue
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Alcaraz J, Nelson CM, Bissell MJ. Biomechanical approaches for studying integration of tissue structure and function in mammary epithelia. J Mammary Gland Biol Neoplasia 2004; 9:361-74. [PMID: 15838605 PMCID: PMC2933199 DOI: 10.1007/s10911-004-1406-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The structure and function of each individual mammary epithelial cell (MEC) is largely controlled by a bidirectional interchange of chemical and mechanical signals with the microenvironment. Most of these signals are tissue-specific, since they arise from the three-dimensional (3D) tissue organization and are modulated during mammary gland development, maturation, pregnancy, lactation, and involution. Although the important role played by structural and mechanical signals in mammary cell and tissue function is being increasingly recognized, quantitative biomechanical approaches are still scarce. Here we review currently available biomechanical tools that allow quantitative examination of individual cells, groups of cells or full monolayers in two-dimensional cultures, and cells in 3D cultures. Current technological limitations and challenges are discussed, with special emphasis on their potential applications in MEC biology. We argue that the combination of biomechanical tools with current efforts in mathematical modeling and in cell and molecular biology applied to 3D cultures provides a powerful approach to unravel the complexity of tissue-specific structure-function relationships.
Collapse
Affiliation(s)
- Jordi Alcaraz
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
- To whom correspondence should be addressed at Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 83-101, Berkeley, California 94720; ;
| | - Celeste M. Nelson
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Mina J. Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
- To whom correspondence should be addressed at Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 83-101, Berkeley, California 94720; ;
| |
Collapse
|