140 results on '"Christine J. Watson"'
Search Results
2. Single-cell lineage tracing in the mammary gland reveals stochastic clonal dispersion of stem/progenitor cell progeny
- Author
-
Felicity M. Davis, Bethan Lloyd-Lewis, Olivia B. Harris, Sarah Kozar, Douglas J. Winton, Leila Muresan, and Christine J. Watson
- Subjects
Science - Abstract
The identity and origin of adult mammary stem cells has been much debated. Here, the authors use a stochastic genetic labelling approach, together with optical tissue clearing, to visualize clonal progeny and show that unipotent stem/progenitor cells contribute to adult mammary gland development.
- Published
- 2016
- Full Text
- View/download PDF
3. The KRAB Zinc Finger Protein Roma/Zfp157 Is a Critical Regulator of Cell-Cycle Progression and Genomic Stability
- Author
-
Teresa L.F. Ho, Guillaume Guilbaud, J. Julian Blow, Julian E. Sale, and Christine J. Watson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Regulation of DNA replication and cell division is essential for tissue growth and maintenance of genomic integrity and is particularly important in tissues that undergo continuous regeneration such as mammary glands. We have previously shown that disruption of the KRAB-domain zinc finger protein Roma/Zfp157 results in hyperproliferation of mammary epithelial cells (MECs) during pregnancy. Here, we delineate the mechanism by which Roma engenders this phenotype. Ablation of Roma in MECs leads to unscheduled proliferation, replication stress, DNA damage, and genomic instability. Furthermore, mouse embryonic fibroblasts (MEFs) depleted for Roma exhibit downregulation of p21Cip1 and geminin and have accelerated replication fork velocities, which is accompanied by a high rate of mitotic errors and polyploidy. In contrast, overexpression of Roma in MECs halts cell-cycle progression, whereas siRNA-mediated p21Cip1 knockdown ameliorates, in part, this phenotype. Thus, Roma is an essential regulator of the cell cycle and is required to maintain genomic stability.
- Published
- 2016
- Full Text
- View/download PDF
4. The Multifaceted Role of STAT3 in Mammary Gland Involution and Breast Cancer
- Author
-
Katherine Hughes and Christine J. Watson
- Subjects
4T1 ,breast cancer ,chitinase 3-like 1 ,CLCA ,cow ,involution ,lysosome ,mammary gland ,microenvironment ,STAT3 ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Since seminal descriptions of signal transducer and activator of transcription 3 (STAT3) as a signal transducer and transcriptional regulator, which is most usually activated by phosphorylation of a specific tyrosine residue, a staggering wealth of research has delineated the key role of this transcription factor as a mediator of mammary gland postlactational regression (involution), and paradoxically, a pro-survival factor in breast cancer and some breast cancer cell lines. STAT3 is a critical regulator of lysosomal-mediated programmed cell death (LM-PCD) during mammary gland involution, where uptake of milk fat globules, and consequent high levels of free fatty acids, cause permeabilisation of lysosomal vesicle membranes, in turn leading to cathepsin protease leakage and cell death. A recent proteomic screen of STAT3-induced changes in lysosomal membrane protein components has highlighted wide-ranging effects of STAT3, which may coordinate LM-PCD via the stimulation of endocytosis, intracellular trafficking, and lysosome biogenesis. In parallel, STAT3 regulates the acute phase response during the first phase of involution, and it contributes to shaping the pro-tumourigenic ‘wound healing’ signature of the gland during the second phase of this process. STAT3 activation during involution is important across species, although some differences exist in the progression of involution in dairy cows. In breast cancer, a number of upstream regulators can lead to STAT3 activation and the effects of phosphorylation of STAT3 are equally wide-ranging. Recent studies have implicated microRNAs in some regulatory pathways. In this review, we will examine the multifaceted role of STAT3 in mammary gland involution and tumourigenesis, incorporating a review of these fundamental processes in tandem with a discussion of recent developments in this field.
- Published
- 2018
- Full Text
- View/download PDF
5. Remembering apoptosis pioneer Andrew Wyllie (1944–2022)
- Author
-
Christine J. Watson, Gerry Melino, and Seamus J. Martin
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry - Published
- 2022
6. The ever‐expanding landscape of cancer therapeutic approaches
- Author
-
Christine J. Watson
- Subjects
medicine.medical_specialty ,Glycosylation ,business.industry ,Cancer ,Cell Biology ,Disease ,Precision medicine ,medicine.disease ,Biochemistry ,RNA Precursors ,medicine ,Humans ,Oncolytic Virus Therapy ,Calcium ,Intensive care medicine ,business ,Molecular Biology - Abstract
Cancer is a leading cause of death and a major health problem worldwide, particularly in more developed countries. There is, therefore, an urgent clinical need to develop more effective therapies to treat cancer and metastatic disease. In this Editorial, the content of The FEBS Journal's Special Issue on Cancer Therapeutics is outlined. The interesting collection of recent articles in this issue covers a wide repertoire of cancer therapeutic approaches. While some of the articles discuss broad-spectrum applications such as immunotherapy and oncolytic virus therapy, others focus on a particular type of cancer or a signalling pathway that has gone awry such as aberrant Ca2+ signalling, glycosylation or pre-mRNA processing. Finally, an article featured in this issue reviews our current understanding of how cancer cells can become dormant, often for decades, and which pathways reactivate these cells to cause relapse. I am sure there is something for everyone in this issue.
- Published
- 2021
7. In conversation with Christine Watson
- Author
-
Paraminder Dhillon and Christine J. Watson
- Subjects
Lineage commitment ,Milk protein ,Watson ,media_common.quotation_subject ,Group leader ,Cell Biology ,Bachelor ,Biochemistry ,Mammary gland development ,Conversation ,Cancer biology ,Molecular Biology ,Classics ,media_common - Abstract
Christine J. Watson is Professor of Cell and Cancer Biology at the University of Cambridge. Christine obtained her Bachelor's (honors) degree in Biochemistry at the University of Glasgow in 1975 and, after a soujourn in Glauco Tocchini-Valentini's lab at the Institute of Cell Biology, Consiglio Nazionale delle Ricerche in Rome, she undertook a PhD in Molecular Genetics at Imperial College London. During her PhD, she looked at differences in gene expression between differentiated and undifferentiated embryonal carcinoma stem cells, inspiring an early interest in gene expression and cell fate determination. Between 1986 and 1992, Christine undertook three postdoctoral research positions that took her from London back to Scotland, where she was first introduced to mammary gland biology through her work with John Clark at the Roslin Institute in Edinburgh. During her time in the Clark lab, Christine identified a factor - later shown to be STAT5 - that binds to the promoter of the milk protein gene β-lactoglobulin. This prompted further work identifying the key role played by the STAT family of transcription factors in mammary gland development. Shortly afterwards, Christine became a group leader at the Roslin Institute and later relocated to the University of Edinburgh to collaborate with Andrew Wyllie. This led to her recruitment to the University of Cambridge in 1998, where she has remained to date. Over the last two decades, the Watson lab has focused on elucidating the mechanisms underlying lineage commitment of mammary stem and progenitor cells and the regulation of cell death in involuting mammary gland. In this interview, Christine discusses her research highlights and provides a glimpse into her personal interests, as she moves towards retirement.
- Published
- 2021
8. Alveolar cells in the mammary gland: lineage commitment and cell death
- Author
-
Christine J, Watson
- Subjects
Mice ,Mammary Glands, Animal ,Cell Death ,Pregnancy ,Alveolar Epithelial Cells ,Animals ,Lactation ,Apoptosis ,Epithelial Cells ,Female - Abstract
The mammary gland provides a spectacular example of physiological cell death whereby the cells that produce milk during lactation are removed swiftly, efficiently, and without inducing inflammation upon the cessation of lactation. The milk-producing cells arise primarily during pregnancy and comprise the alveolar lineage that is specified by signalling pathways and factors that are activated in response to pregnancy hormones. There are at least two alveolar sub-lineages, one of which is marked by the presence of binucleate cells that are especially susceptible to programmed cell death during involution. This process of post-lactational regression, or involution, is carefully orchestrated and occurs in two phases, the first results in a rapid switch in cell fate with the secretory epithelial cells becoming phagocytes whereupon they destroy dead and dying cells from milk. This reversible phase is followed by the second phase that is marked by an influx of immune cells and a remodelling of the gland to replace the alveolar cells with re-differentiated adipocytes, resulting in a return to the pre-pregnant state in preparation for any subsequent pregnancies. The mouse mammary gland provides an excellent experimental tool with which to investigate lineage commitment and the mechanisms of programmed cell death that occur in a normal physiological process. Importantly, involution has highlighted a role for lysoptosis, a mechanism of cell death that is mediated by lysosomal cathepsins and their endogenous inhibitors, serpins. In this review, I discuss alveolar lineage commitment during pregnancy and the programmed cell death pathways that destroy these cells during involution.
- Published
- 2022
9. The immune environment of the mammary gland fluctuates during post-lactational regression and correlates with tumour growth rate
- Author
-
Jessica Hitchcock, Katherine Hughes, Sara Pensa, Bethan Lloyd-Lewis, and Christine J. Watson
- Subjects
Involution ,Mouse ,Mammary gland ,Immune cells ,Postpartum Period ,Breast Neoplasms ,Tumourigenesis ,Mice ,Mammary Glands, Animal ,Pregnancy ,Animals ,Humans ,Lactation ,Female ,Mammary Glands, Human ,Molecular Biology ,Developmental Biology ,Aged - Abstract
Post-lactational mammary gland regression encompasses extensive programmed cell death and removal of milk-producing epithelial cells, breakdown of extracellular matrix components and redifferentiation of stromal adipocytes. This highly regulated involution process is associated with a transient increased risk of breast cancer in women. Using a syngeneic tumour model, we show that tumour growth is significantly altered depending on the stage of involution at which tumour cells are implanted. Tumour cells injected at day 3 involution grew faster than those in nulliparous mice, whereas tumours initiated at day 6 involution grew significantly slower. These differences in tumour progression correlate with distinct changes in innate immune cells, in particular among F4/80-expressing macrophages and among TCRδ+ unconventional T cells. Breast cancer post-pregnancy risk is exacerbated in older first-time mothers and, in our model, initial tumour growth is moderately faster in aged mice compared with young mice. Our results have implications for breast cancer risk and the use of anti-inflammatory therapeutics for postpartum breast cancers.
- Published
- 2021
10. A pilot study evaluating GSK1070806 inhibition of interleukin-18 in renal transplant delayed graft function
- Author
-
H. A. Stirnadel-Farrant, Mary Birchler, R. Schroyer, Christine J. Watson, Oriol Bestard, Neil S. Sheerin, Stephen DeWall, John D. Scott, Stephen J. Wigmore, Lee Abberley, R. Noble, Ewen M Harrison, Menna R. Clatworthy, Robert B. Kirkpatrick, Luke Devey, M. Busz, David B. Kingsmore, Katrina Stevenson, S. Andrews, K. S. Thorneloe, E. Wlodek, Alexander N.R. Weber, D. Krull, Kirkpatrick, R. B. [0000-0002-8671-2361], Andrews, S. [0000-0002-5407-7201], Thorneloe, K. S. [0000-0003-3640-9839], Weber, A. [0000-0001-6952-5081], Apollo - University of Cambridge Repository, Kirkpatrick, RB [0000-0002-8671-2361], Andrews, S [0000-0002-5407-7201], Thorneloe, KS [0000-0003-3640-9839], and Weber, A [0000-0001-6952-5081]
- Subjects
Male ,Hyperkalemia ,Physiology ,medicine.medical_treatment ,Biopsy ,030232 urology & nephrology ,Trasplantament renal ,Pilot Projects ,030204 cardiovascular system & hematology ,Urine ,Biochemistry ,Kidney transplantation ,0302 clinical medicine ,Clinical endpoint ,Renal Transplantation ,Medicine ,education.field_of_study ,Multidisciplinary ,Acute kidney injury ,Interleukin-18 ,Diàlisi ,Acute Kidney Injury ,Middle Aged ,Tissue Donors ,Body Fluids ,Nephrology ,Female ,medicine.symptom ,Anatomy ,Biòpsia ,Research Article ,Adult ,medicine.medical_specialty ,Science ,Population ,Urology ,Delayed Graft Function ,Surgical and Invasive Medical Procedures ,Antibodies, Monoclonal, Humanized ,Urinary System Procedures ,03 medical and health sciences ,Medical Dialysis ,Humans ,Adverse effect ,education ,Immunohistochemistry Techniques ,Dialysis ,Aged ,Medicine and health sciences ,Transplantation ,Biology and life sciences ,business.industry ,Proteins ,Kidneys ,Organ Transplantation ,Renal System ,medicine.disease ,Kidney Transplantation ,Histochemistry and Cytochemistry Techniques ,Research and analysis methods ,Reperfusion ,Immunologic Techniques ,Interferons ,business ,Physiological Processes - Abstract
Funder: GlaxoSmithKline; funder-id: http://dx.doi.org/10.13039/100004330, Introduction: Delayed graft function (DGF) following renal transplantation is a manifestation of acute kidney injury (AKI) leading to poor long-term outcome. Current treatments have limited effectiveness in preventing DGF. Interleukin-18 (IL18), a biomarker of AKI, induces interferon-γ expression and immune activation. GSK1070806, an anti-IL18 monoclonal antibody, neutralizes activated (mature) IL18 released from damaged cells following inflammasome activation. This phase IIa, single-arm trial assessed the effect of a single dose of GSK1070806 on DGF occurrence post donation after circulatory death (DCD) kidney transplantation. Methods: The 3 mg/kg intravenous dose was selected based on prior studies and physiologically based pharmacokinetic (PBPK) modeling, indicating the high likelihood of a rapid and high level of IL18 target engagement when administered prior to kidney allograft reperfusion. Utilization of a Bayesian sequential design with a background standard-of-care DGF rate of 50% based on literature, and confirmed via extensive registry data analyses, enabled a statistical efficacy assessment with a minimal sample size. The primary endpoint was DGF frequency, defined as dialysis requirement ≤7 days post transplantation (except for hyperkalemia). Secondary endpoints included safety, pharmacokinetics and pharmacodynamic biomarkers. Results: GSK1070806 administration was associated with IL18-GSK1070806 complex detection and increased total serum IL18 levels due to IL18 half-life prolongation induced by GSK1070806 binding. Interferon-γ−induced chemokine levels declined or remained unchanged in most patients. Although the study was concluded prior to the Bayesian-defined stopping point, 4/7 enrolled patients (57%) had DGF, exceeding the 50% standard-of-care rate, and an additional two patients, although not reaching the protocol-defined DGF definition, demonstrated poor graft function. Six of seven patients experienced serious adverse events (SAEs), including two treatment-related SAEs. Conclusion: Overall, using a Bayesian design and extensive PBPK dose modeling with only a small sample size, it was deemed unlikely that GSK1070806 would be efficacious in preventing DGF in the enrolled DCD transplant population. Trial registration: NCT02723786.
- Published
- 2021
- Full Text
- View/download PDF
11. How should we define mammary stem cells?
- Author
-
Christine J. Watson
- Subjects
0303 health sciences ,Stem Cells ,Cell ,Cell Differentiation ,Cell Biology ,Biology ,Cell biology ,Transplantation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine.anatomical_structure ,Mammary Glands, Animal ,Live cell imaging ,medicine ,Animals ,Cell Lineage ,Epigenetics ,Progenitor cell ,Stem cell ,Reprogramming ,030217 neurology & neurosurgery ,030304 developmental biology ,Progenitor - Abstract
Mammary stem cells (MaSCs) have been defined by cell surface marker expression and their ability to repopulate a cleared fat pad, a capacity now known to result from reprogramming upon transplantation. Furthermore, lineage-tracing studies have provoked controversy as to whether MaSCs are unipotent or bi/multipotent. Various innovative experimental approaches, including single-cell RNA sequencing (scRNA-Seq), epigenetic analyses, deep tissue and live imaging, and advanced mouse models, have provided new and unexpected insights into stem and progenitor cells; thus, it is now timely to reappraise our concept of the MaSC hierarchy. Here, I highlight misconceptions, suggest definitions of stem and progenitor cells, and propose a way forward in our search for an understanding of MaSCs.
- Published
- 2020
12. Mammary development in the embryo and adult: new insights into the journey of morphogenesis and commitment
- Author
-
Christine J. Watson, Walid T. Khaled, Watson, Christine J [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
Organogenesis ,Mammary gland ,Cell ,Morphogenesis ,Embryonic Development ,Computational biology ,Biology ,Lineage specification ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mammary Glands, Animal ,medicine ,Animals ,Humans ,Cell Lineage ,Microbiome ,Progenitor cell ,Mammary Glands, Human ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Stem Cells ,Embryo ,Cell Differentiation ,Epithelial Cells ,Embryo, Mammalian ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Stem cell ,Function (biology) ,Developmental Biology - Abstract
The mammary gland is a unique tissue and the defining feature of the class Mammalia. It is a late-evolving epidermal appendage that has the primary function of providing nutrition for the young, although recent studies have highlighted additional benefits of milk including the provision of passive immunity and a microbiome and, in humans, the psychosocial benefits of breastfeeding. In this Review, we outline the various stages of mammary gland development in the mouse, with a particular focus on lineage specification and the new insights that have been gained by the application of recent technological advances in imaging in both real-time and three-dimensions, and in single cell RNA sequencing. These studies have revealed the complexity of subpopulations of cells that contribute to the mammary stem and progenitor cell hierarchy and we suggest a new terminology to distinguish these cells.
- Published
- 2020
- Full Text
- View/download PDF
13. The spectrum of STAT functions in mammary gland development
- Author
-
Katherine Hughes, Christine J. Watson, Hughes, Katherine [0000-0002-3331-1249], Watson, Christine [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
medicine.medical_specialty ,mammary gland ,Mammary gland ,Review ,gene targeting ,Alveolar cells ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Involution (medicine) ,STAT1 ,STAT3 ,mouse ,030304 developmental biology ,STAT6 ,0303 health sciences ,biology ,STAT ,apoptosis ,General Medicine ,differentiation ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,030220 oncology & carcinogenesis ,STAT protein ,biology.protein ,Stem cell ,microarray - Abstract
The signal transducer and activator of transcription (STAT) family of transcription factors have a spectrum of functions in mammary gland development. In some cases these roles parallel those of STATs in other organ systems, while in other instances the function of individual STATs in the mammary gland is specific to this tissue. In the immune system, STAT6 is associated with differentiation of T helper cells, while in the mammary gland, it has a fundamental role in the commitment of luminal epithelial cells to the alveolar lineage. STAT5A is required for the production of luminal progenitor cells from mammary stem cells and is essential for the differentiation of milk producing alveolar cells during pregnancy. By contrast, the initiation of regression following weaning heralds a dramatic and specific activation of STAT3, reflecting its pivotal role in the regulation of cell death and tissue remodeling during mammary involution. Although it has been demonstrated that STAT1 is regulated during a mammary developmental cycle, it is not yet determined whether it has a specific, non-redundant function. Thus, the mammary gland constitutes an unusual example of an adult organ in which different STATs are sequentially activated to orchestrate the processes of functional differentiation, cell death and tissue remodeling.
- Published
- 2020
- Full Text
- View/download PDF
14. Sinus‐like dilatations of the mammary milk ducts, Ki67 expression, and <scp>CD</scp> 3‐positive T lymphocyte infiltration, in the mammary gland of wild European rabbits during pregnancy and lactation
- Author
-
Christine J. Watson, Katherine Hughes, Hughes, Katherine [0000-0002-3331-1249], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,mammary gland ,Pathology ,medicine.medical_specialty ,Histology ,T-Lymphocytes ,CD3 ,Mammary gland ,rabbit ,lactation ,Biology ,Brief Communication ,mastitis ,03 medical and health sciences ,Cytokeratin ,Mammary Glands, Animal ,Immune system ,Pregnancy ,Lactation ,medicine ,Animals ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Innate immune system ,tumour ,milk duct ,Epithelial Cells ,Cell Biology ,T lymphocyte ,Epithelium ,Ki-67 Antigen ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Pregnancy, Animal ,sinus ,Female ,Rabbits ,Anatomy ,Developmental Biology - Abstract
Sinus‐like dilatations of the mammary duct are recognisable in the mammary gland of pregnant and lactating wild European rabbits. These dilatations exhibit a bilaminar epithelial lining, with luminal epithelial cells expressing basal and lateral E‐cadherin. Occasional binucleated mammary epithelial cells are present in the luminal layer. Underlying the luminal epithelial cells is a basal layer of cytokeratin 14‐positive cells, supported by a thin layer of fibrous tissue. Multi‐segmental epithelial proliferation, as indicated by Ki67 expression, is apparent in the luminal epithelial cells, suggesting a capacity for division during pregnancy and lactation. CD3‐positive T lymphocytes are present both intraepithelially, suggesting exocytosis, and in foci subjacent to the ductular epithelium. We consider that sinus‐like dilatations of the mammary duct may have the potential to give rise to a subset of the mammary gland neoplasms classified as ductal in origin. Milk accumulation in these sinus‐like dilatations is likely to provide a niche for bacterial replication in cases of mastitis in rabbits. These structures are an important component of the innate immune system of the mammary gland, both as a physical barrier and as an interface between the milk and mammary immune cells.
- Published
- 2018
15. A multifunctional 3D co-culture system for studies of mammary tissue morphogenesis and stem cell biology.
- Author
-
Jonathan J Campbell, Natalia Davidenko, Maria M Caffarel, Ruth E Cameron, and Christine J Watson
- Subjects
Medicine ,Science - Abstract
Studies on the stem cell niche and the efficacy of cancer therapeutics require complex multicellular structures and interactions between different cell types and extracellular matrix (ECM) in three dimensional (3D) space. We have engineered a 3D in vitro model of mammary gland that encompasses a defined, porous collagen/hyaluronic acid (HA) scaffold forming a physiologically relevant foundation for epithelial and adipocyte co-culture. Polarized ductal and acinar structures form within this scaffold recapitulating normal tissue morphology in the absence of reconstituted basement membrane (rBM) hydrogel. Furthermore, organoid developmental outcome can be controlled by the ratio of collagen to HA, with a higher HA concentration favouring acinar morphological development. Importantly, this culture system recapitulates the stem cell niche as primary mammary stem cells form complex organoids, emphasising the utility of this approach for developmental and tumorigenic studies using genetically altered animals or human biopsy material, and for screening cancer therapeutics for personalised medicine.
- Published
- 2011
- Full Text
- View/download PDF
16. Mammary Stem Cells: Premise, Properties, and Perspectives
- Author
-
Felicity M. Davis, Olivia B Harris, Christine J Watson, and Bethan Lloyd-Lewis
- Subjects
0301 basic medicine ,Cellular differentiation ,Mammary gland ,Cell lineage ,Biology ,Mammary gland development ,Mice ,03 medical and health sciences ,Mammary Glands, Animal ,Cancer stem cell ,Lineage tracing ,medicine ,Animals ,Humans ,Cell Lineage ,Stem Cell Niche ,Mammary Glands, Human ,Stem Cells ,Cell Differentiation ,Cell Biology ,Stem cell niche ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Female ,Stem cell ,Neuroscience - Abstract
Adult mammary stem cells (MaSCs) drive postnatal organogenesis and remodeling in the mammary gland, and their longevity and potential have important implications for breast cancer. However, despite intense investigation the identity, location, and differentiation potential of MaSCs remain subject to deliberation. The application of genetic lineage-tracing models, combined with quantitative 3D imaging and biophysical methods, has provided new insights into the mammary epithelial hierarchy that challenge classical definitions of MaSC potency and behaviors. We review here recent advances - discussing fundamental unresolved properties of MaSC potency, dynamics, and plasticity - and point to evolving technologies that promise to shed new light on this intractable debate. Elucidation of the physiological mammary differentiation hierarchy is paramount to understanding the complex heterogeneous breast cancer landscape.
- Published
- 2017
17. Dynamic architectural interplay between leucocytes and mammary epithelial cells
- Author
-
Katherine Hughes, Jessica R. Hitchcock, Christine J. Watson, Olivia B Harris, Hughes, Katherine [0000-0002-3331-1249], Watson, Christine J [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,mammary gland ,Stromal cell ,Mammary gland ,Biology ,Biochemistry ,Alveolar cells ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,leucocytes ,medicine ,involution ,Leukocytes ,Animals ,Humans ,Lactation ,Involution (medicine) ,Mammary Glands, Human ,Molecular Biology ,Mice, Inbred BALB C ,Myoepithelial cell ,imaging ,Epithelial Cells ,Cell Biology ,Epithelium ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Mammary Epithelium ,030220 oncology & carcinogenesis ,Female - Abstract
The adult mammary gland undergoes dynamic changes during puberty and the postnatal developmental cycle. The mammary epithelium is composed of a bilayer of outer basal, or myoepithelial, cells and inner luminal cells, the latter lineage giving rise to the milk-producing alveolar cells during pregnancy. These luminal alveolar cells undergo Stat3-mediated programmed cell death following the cessation of lactation. It is established that immune cells in the microenvironment of the gland have a role to play both in the ductal outgrowth during puberty and in the removal of dead cells and remodelling of the stroma during the process of postlactational regression. However, most studies have focussed on the role of the stromal immune cell compartment or have quantified immune cell populations in tissue extracts. Our recent development of protocols for deep imaging of the mammary gland in three dimensions (3D) has enabled the architectural relationship between immune cells and the epithelium to be examined in detail, and we have discovered a surprisingly dynamic relationship between the basal epithelium and leucocytes. Furthermore, we have observed morphological changes in the myoepithelial cells, as involution progresses, which were not revealed by previous work in 2D tissue sections and whole tissue. This dynamic architecture suggests a role for myoepithelial cells in the orderly progression of involution. We conclude that deep imaging of mammary gland and other tissues is essential for analysing complex interactions between cellular compartments.
- Published
- 2019
18. The Mammary Microenvironment in Mastitis in Humans, Dairy Ruminants, Rabbits and Rodents: A One Health Focus
- Author
-
Katherine Hughes, Christine J. Watson, Hughes, Katherine [0000-0002-3331-1249], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cancer Research ,Microenvironment ,Ruminant ,Mammary gland ,Rodentia ,Mastitis ,Rabbit ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Mammary Glands, Animal ,medicine ,Animals ,Humans ,Involution (medicine) ,Subclinical mastitis ,One Health ,Pregnancy ,Sheep ,Laboratory Animal Models ,Ruminants ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cellular Microenvironment ,030220 oncology & carcinogenesis ,Immunology ,Cattle ,Female ,Rabbits - Abstract
The One Health concept promotes integrated evaluation of human, animal, and environmental health questions to expedite advances benefiting all species. A recognition of the multi-species impact of mastitis as a painful condition with welfare implications leads us to suggest that mastitis is an ideal target for a One Health approach. In this review, we will evaluate the role of the mammary microenvironment in mastitis in humans, ruminants and rabbits, where appropriate also drawing on studies utilising laboratory animal models. We will examine subclinical mastitis, clinical lactational mastitis, and involution-associated, or dry period, mastitis, highlighting important anatomical and immunological species differences. We will synthesise knowledge gained across different species, comparing and contrasting disease presentation. Subclinical mastitis (SCM) is characterised by elevated Na/K ratio, and increased milk IL-8 concentrations. SCM affecting the breastfeeding mother may result in modulation of infant mucosal immune system development, whilst in ruminants notable milk production losses may ensue. In the case of clinical lactational mastitis, we will focus on mastitis caused by Staphylococcus aureus and Escherichia coli. Understanding of the pathogenesis of involution-associated mastitis requires characterization of the structural and molecular changes occurring during involution and we will review these changes across species. We speculate that milk accumulation may act as a nidus for infection, and that the involution 'wound healing phenotype' may render the tissue susceptible to bacterial infection. We will discuss the impact of concurrent pregnancy and a 'parallel pregnancy and involution signature' during bovine mammary involution.
- Published
- 2018
- Full Text
- View/download PDF
19. An Engineered Human Adipose/Collagen Model for In Vitro Breast Cancer Cell Migration Studies
- Author
-
Lorraine S. Berry, Ruth E. Cameron, Jenny Gomm, Robert D. Hume, Stefanie Reichelt, Christine J. Watson, and Michael E. D’Angelo
- Subjects
0301 basic medicine ,Stromal cell ,Biomedical Engineering ,Adipose tissue ,Bioengineering ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Biochemistry ,Models, Biological ,Collagen Type I ,Biomaterials ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Tissue engineering ,Cell Movement ,medicine ,Adipocytes ,Tumor Cells, Cultured ,Humans ,skin and connective tissue diseases ,Cell Line, Transformed ,Tissue Engineering ,Tissue Scaffolds ,Mesenchymal stem cell ,Cancer ,Mesenchymal Stem Cells ,medicine.disease ,030104 developmental biology ,Adipose Tissue ,Adipogenesis ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis - Abstract
Adipocytes are one of the major stromal cell components of the human breast. These cells play a key role in the development of the gland and are implicated in breast tumorigenesis. Frequently, directional stromal collagen I fibers are found surrounding aggressive breast tumors. These fibers enhance breast cancer cell migration and are associated with poor patient prognosis. We sought to recapitulate these stromal components in vitro to provide a three-dimensional (3D) model comprising human adipose tissue and anisotropic collagen fibers. We developed a human mesenchymal stem cell (hMSC) cell line capable of undergoing differentiation into mature adipocytes by immortalizing hMSCs, isolated from breast reduction mammoplasties, through retroviral transduction. These immortalized hMSCs were seeded in engineered collagen I scaffolds with directional internal architecture, and adipogenesis was chemically induced, resulting in human adipose tissue being synthesized in vitro in an architectural structure associated with breast tumorigenesis. Subsequently, fluorescently labeled cells from an established breast cancer cell line were seeded into this model, cocultured for 7 days and imaged using multiphoton microscopy. Enhanced breast cancer cell migration was observed in the adipose-containing model over empty scaffold controls, demonstrating an adipocyte-mediated influence on breast cancer cell migration. Thus, this 3D in vitro model recapitulates the migratory effects of adipocytes observed on breast cancer cells and suggests that it could have utility with fresh breast tumor biopsies as an assay for cancer therapeutic efficacy in personalized medicine strategies.
- Published
- 2018
20. Stat3-mediated alterations in lysosomal membrane protein composition
- Author
-
Bethan, Lloyd-Lewis, Caroline C, Krueger, Timothy J, Sargeant, Michael E, D'Angelo, Michael J, Deery, Renata, Feret, Julie A, Howard, Kathryn S, Lilley, and Christine J, Watson
- Subjects
Proteomics ,STAT3 Transcription Factor ,mammary gland ,Cell Death ,Proteome ,Genomics and Proteomics ,Lysosome-Associated Membrane Glycoproteins ,Epithelial Cells ,Intracellular Membranes ,STAT3 ,mammary epithelial cells ,Mammary Glands, Animal ,lysosome purification ,lysosome ,involution ,Animals ,Female ,Lysosomes ,Cells, Cultured ,Signal Transduction - Abstract
Lysosome function is essential in cellular homeostasis. In addition to its recycling role, the lysosome has recently been recognized as a cellular signaling hub. We have shown in mammary epithelial cells, both in vivo and in vitro, that signal transducer and activator of transcription 3 (Stat3) modulates lysosome biogenesis and can promote the release of lysosomal proteases that culminates in cell death. To further investigate the impact of Stat3 on lysosomal function, we conducted a proteomic screen of changes in lysosomal membrane protein components induced by Stat3 using an iron nanoparticle enrichment strategy. Our results show that Stat3 activation not only elevates the levels of known membrane proteins but results in the appearance of unexpected factors, including cell surface proteins such as annexins and flotillins. These data suggest that Stat3 may coordinately regulate endocytosis, intracellular trafficking, and lysosome biogenesis to drive lysosome-mediated cell death in mammary epithelial cells. The methodologies described in this study also provide significant improvements to current techniques used for the purification and analysis of the lysosomal proteome.
- Published
- 2018
21. STAT3 the oncogene - still eluding therapy?
- Author
-
Matthew S Wake, Christine J. Watson, Watson, Christine [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
STAT3 Transcription Factor ,Programmed cell death ,medicine.medical_treatment ,Biology ,medicine.disease_cause ,Biochemistry ,stat ,STAT3 ,cancer therapeutics ,Neoplasms ,inhibitors ,medicine ,cancer ,Humans ,Gene silencing ,Molecular Targeted Therapy ,signalling ,Receptor ,Molecular Biology ,Transcription factor ,Oncogenes ,Cell Biology ,Protein Structure, Tertiary ,Gene Expression Regulation, Neoplastic ,Cytokine ,tumourigenesis ,Cancer research ,biology.protein ,Carcinogenesis - Abstract
The STAT family of transcription factors (signal transducers and activators of transcription) transduce signals from cytokine receptors to the nucleus, where STAT dimers bind to DNA and regulate transcription. STAT3 is the most ubiquitous of the STATs, being activated by a wide variety of cytokines and growth factors. STAT3 has many roles in physiological processes such as inflammatory signalling, aerobic glycolysis and immune suppression, and was also the first family member shown to be aberrantly activated in a wide range of both solid and liquid tumours. STAT3 promotes tumorigenesis by regulating the expression of various target genes, including cell-cycle regulators, angiogenic factors and anti-apoptosis genes. Paradoxically, in some circumstances, STAT3 signalling induces cell death. The best known example is the involuting mammary gland, where STAT3 is essential for induction of a lysosomal pathway of cell death. Nevertheless, direct silencing or inhibition of STAT3 diminishes tumour growth and survival in both animal and human studies. This suggests that abolishing STAT3 activity may be an effective cancer therapeutic strategy. However, despite this potential as a therapeutic target, and the extensive attempts by many laboratories and pharmaceutical companies to develop an effective STAT3 inhibitor for use in the clinic, no direct STAT3 inhibitor has been approved for clinical use. In this review, we focus on the role of STAT3 in tumorigenesis, and discuss its potential as a therapeutic target for cancer treatment.
- Published
- 2015
22. Signal transducer and activator of transcription 3 and the phosphatidylinositol 3-kinase regulatory subunits p55α and p50α regulate autophagyin vivo
- Author
-
Christine J. Watson, Sara Pensa, C. Ronald Kahn, Timothy J. Sargeant, Henrike K. Resemann, and Bethan Lloyd-Lewis
- Subjects
Programmed cell death ,Autophagy ,Cell Biology ,Biology ,STAT3 Transcription Factor ,BAG3 ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,Lysosome ,medicine ,biology.protein ,Involution (medicine) ,STAT3 ,Molecular Biology ,Mammary gland involution - Abstract
Mammary gland involution involves a process that includes one of the most dramatic examples of cell death in an adult mammalian organism. We have previously shown that signal transducer and activator of transcription 3 (Stat3) regulates a lysosomal pathway of cell death in the first 48 h of involution and induces lysosome leakiness in mammary epithelial cells. Interestingly, Stat3 is associated also with the striking induction of autophagy that occurs concomitantly with cell death, presumably as a transient survival mechanism. The phosphatidylinositol 3-kinase regulatory subunits p55α and p50α are dramatically and specifically upregulated at the transcriptional level by Stat3 at the onset of involution. We show here that ablation of either Stat3 or p55α/p50α in vivo affects autophagy during involution. We used two different cell culture models (normal mammary epithelial cells and mouse embryonic fibroblasts) to further investigate the role of p55α/p50α in autophagy regulation. Our results demonstrate a direct role for p55α/p50α as inhibitors of autophagy mediated by p85α. Thus, Stat3 and its downstream targets p55α/p50α are key regulators of the balance between autophagy and cell death in vivo.
- Published
- 2014
23. The PI3K regulatory subunits p55α and p50α regulate cell death in vivo
- Author
-
Thomas Reinheckel, NJ Clarke, CR Kahn, Henrike K. Resemann, Sara Pensa, Christine J. Watson, Kathrine Abell, Kevin Neoh, Peter A. Kreuzaler, Watson, Christine [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
Male ,Mice, Knockout ,Cathepsin ,Original Paper ,Programmed cell death ,Cell Death ,Cell growth ,Protein subunit ,education ,Cell Biology ,Biology ,Molecular biology ,Cell biology ,Cathepsin L ,Mice ,Phosphatidylinositol 3-Kinases ,Protein Subunits ,biology.protein ,Animals ,Female ,Involution (medicine) ,STAT3 ,Molecular Biology ,Mammary gland involution - Abstract
The phosphatidylinositol 3-kinase (PI3K) regulatory subunits p55α and p50α are coordinately transcriptionally upregulated by signal transducer and activator of transcription 3 (Stat3) at the onset of mammary gland involution, a process that requires Stat3. Deletion of both p55α and p50α subunits in vivo abrogated mammary epithelial cell death during involution. This was associated also with reduced cytosolic levels and activity of the cysteine protease cathepsin L, which is implicated in lysosomal-mediated programmed cell death (LM-PCD) and is upregulated in involution. Furthermore, involution is delayed in cathepsin L-deficient mice suggesting that the p55α/p50α subunits mediate cell death in part by elevating the level of cathepsin L resulting in increased cytosolic activity. Surprisingly, we found that p55α/p50α localize to the nucleus where they bind to chromatin and regulate transcription of a subset of inflammatory/acute phase genes that are also Stat3 targets. Our findings reveal a novel role for these PI3K regulatory subunits as regulators of LM-PCD in vivo.
- Published
- 2014
24. The KRAB domain zinc finger protein, Zfp157, is expressed in multiple tissues during mouse embryogenesis and in specific cells in adult mammary gland and skin
- Author
-
Christine J. Watson, Carrie H. Oliver, and Jennifer Nichols
- Subjects
Zinc finger ,medicine.medical_specialty ,Reporter gene ,Mammary gland ,Ovary ,Cell Biology ,Biology ,Hair follicle ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,Epiblast ,Internal medicine ,Genetics ,medicine ,Transcription factor ,STAT6 - Abstract
The functions of members of the large family of transcriptional repressors, the KRAB domain zinc finger proteins, are not well described. We have identified a new member of this family, Zfp157, as a downstream target of the transcription factor Stat6 in mammary gland. Using a gene-trap approach, we have generated mice harboring a Zfp157-LacZ reporter gene. We have characterized the expression of this reporter during mouse embryogenesis and show that it is expressed in the epiblast and subsequently in a number of embryonic tissues including brain, ovary, intestine, kidney, lung, mammary gland, and hair follicle. In the adult, Zfp157 continues to be expressed in a wide range of tissues while specific patterns of reporter gene expression are apparent in the mammary gland, primarily in the basal epithelial cells of ducts and in the sebaceous glands of hair follicles. These data lay the foundation for further work on the function of Zfp157. genesis 51:179–186, 2013. © 2013 Wiley Periodicals, Inc.
- Published
- 2013
25. Analysis of the Involuting Mouse Mammary Gland: An In Vivo Model for Cell Death
- Author
-
Bethan, Lloyd-Lewis, Timothy J, Sargeant, Peter A, Kreuzaler, Henrike K, Resemann, Sara, Pensa, and Christine J, Watson
- Subjects
Mice ,Mammary Glands, Animal ,Cell Death ,Transcription, Genetic ,Pregnancy ,Animals ,Lactation ,Epithelial Cells ,Female ,Lysosomes ,Signal Transduction - Abstract
Involution of the mammary gland occurs at the end of every period of lactation and is an essential process to return the gland to a pre-pregnant state in readiness for the next pregnancy. Involution is a complex process of regulated alveolar cell death coupled with tissue remodeling and requires exquisite control of transcription and signaling. These processes can be investigated using a variety of molecular and morphological approaches.In this chapter we describe how to initiate involution and collect mammary glands, measure involution morphologically, and quantify lysosomal leakiness in mammary tissue and in cultured mammary epithelial cells. These procedures encompass a range of microscopy and molecular biology techniques.
- Published
- 2016
26. Analysis of the Involuting Mouse Mammary Gland: An In Vivo Model for Cell Death
- Author
-
Christine J. Watson, Peter A. Kreuzaler, Sara Pensa, Timothy J. Sargeant, Henrike K. Resemann, and Bethan Lloyd-Lewis
- Subjects
0301 basic medicine ,Programmed cell death ,Mammary gland ,Caspase 3 ,Biology ,Cell biology ,Alveolar cells ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,In vivo ,Lysosome ,Lactation ,medicine ,Involution (medicine) - Abstract
Involution of the mammary gland occurs at the end of every period of lactation and is an essential process to return the gland to a pre-pregnant state in readiness for the next pregnancy. Involution is a complex process of regulated alveolar cell death coupled with tissue remodeling and requires exquisite control of transcription and signaling. These processes can be investigated using a variety of molecular and morphological approaches.In this chapter we describe how to initiate involution and collect mammary glands, measure involution morphologically, and quantify lysosomal leakiness in mammary tissue and in cultured mammary epithelial cells. These procedures encompass a range of microscopy and molecular biology techniques.
- Published
- 2016
27. Imaging the mammary gland and mammary tumours in 3D: optical tissue clearing and immunofluorescence methods
- Author
-
Bethan, Lloyd-Lewis, Felicity M, Davis, Olivia B, Harris, Jessica R, Hitchcock, Filipe C, Lourenco, Mathias, Pasche, and Christine J, Watson
- Subjects
Fluorescence microscopy ,Microscopy, Confocal ,Light sheet fluorescence microscopy ,Optical Imaging ,Mammary gland ,Fluorescent Antibody Technique ,Mammary Neoplasms, Animal ,Mice ,Imaging, Three-Dimensional ,Mammary Glands, Animal ,Breast cancer ,3D imaging ,Animals ,Lactation ,Female ,Two-photon microscopy ,Research Article ,Tissue clearing - Abstract
Background High-resolution 3D imaging of intact tissue facilitates cellular and subcellular analyses of complex structures within their native environment. However, difficulties associated with immunolabelling and imaging fluorescent proteins deep within whole organs have restricted their applications to thin sections or processed tissue preparations, precluding comprehensive and rapid 3D visualisation. Several tissue clearing methods have been established to circumvent issues associated with depth of imaging in opaque specimens. The application of these techniques to study the elaborate architecture of the mouse mammary gland has yet to be investigated. Methods Multiple tissue clearing methods were applied to intact virgin and lactating mammary glands, namely 3D imaging of solvent-cleared organs, see deep brain (seeDB), clear unobstructed brain imaging cocktails (CUBIC) and passive clarity technique. Using confocal, two-photon and light sheet microscopy, their compatibility with whole-mount immunofluorescent labelling and 3D imaging of mammary tissue was examined. In addition, their suitability for the analysis of mouse mammary tumours was also assessed. Results Varying degrees of optical transparency, tissue preservation and fluorescent signal conservation were observed between the different clearing methods. SeeDB and CUBIC protocols were considered superior for volumetric fluorescence imaging and whole-mount histochemical staining, respectively. Techniques were compatible with 3D imaging on a variety of platforms, enabling visualisation of mammary ductal and lobulo-alveolar structures at vastly improved depths in cleared tissue. Conclusions The utility of whole-organ tissue clearing protocols was assessed in the mouse mammary gland. Most methods utilised affordable and widely available reagents, and were compatible with standard confocal microscopy. These techniques enable high-resolution, 3D imaging and phenotyping of mammary cells and tumours in situ, and will significantly enhance our understanding of both normal and pathological mammary gland development. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0754-9) contains supplementary material, which is available to authorized users.
- Published
- 2016
28. Isolation of Mouse Mammary Epithelial Subpopulations: A Comparison of Leading Methods
- Author
-
Geoffrey J. Lindeman, Christine J. Watson, Jane E. Visvader, Joseph L. Regan, John Stingl, Howard Kendrick, Matthew J. Smalley, Michael D. Prater, and Julie Sheridan
- Subjects
Cancer Research ,Staining and Labeling ,Isolation (health care) ,Cellular differentiation ,Mammary cells ,Cell Differentiation ,Cell Separation ,Computational biology ,Biology ,Standard technique ,Epithelium ,Organoids ,Adult Stem Cells ,Mice ,Mammary Glands, Animal ,Oncology ,Antibody Specificity ,Antigens, Surface ,Immunology ,Cell separation ,Animals ,Female ,Progenitor cell ,Cells, Cultured ,Adult stem cell - Abstract
Isolation of mammary epithelial subpopulations, including stem and progenitor cells, has become a standard technique in recent years. However, a number of methods and approaches for this have developed and the relative benefits of the different approaches, and the reason for their development, have not always been clear. Here, three of the leading laboratories working on the separation of mammary cell subpopulations have summarised their methods, highlighted their differences and similarities and also discussed the reasoning behind the approaches they have taken. This article will assist workers establishing mammary cell separation protocols in their laboratories to make informed choices about the methods they should use.
- Published
- 2012
29. Killing a cancer: what are the alternatives?
- Author
-
Christine J. Watson and Peter A. Kreuzaler
- Subjects
Programmed cell death ,Cell Death ,Effector ,Applied Mathematics ,General Mathematics ,Necroptosis ,Autophagy ,Cancer therapy ,Cancer ,Biology ,medicine.disease ,Cell biology ,Necrosis ,The Hallmarks of Cancer ,Neoplasms ,Receptor-Interacting Protein Serine-Threonine Kinases ,medicine ,Humans ,Lysosomes ,Neuroscience ,Signal Transduction - Abstract
Evading programmed cell death is one of the hallmarks of cancer. Conversely, inducing cell death by pharmacological means is the basis of almost every non-invasive cancer therapy. Research over the past decade has greatly increased our understanding of non-apoptotic programmed cell death events, such as lysosomal-mediated cell death, necroptosis and cell death with autophagy. It is becoming clear that an intricate effector network connects many of these classical and non-classical death pathways. In this Review, we discuss converging and diverging features of these pathways, as well as attempts to exploit this newly gained knowledge pharmacologically to provide therapeutics for cancer.
- Published
- 2012
30. Biomimetic collagen scaffolds with anisotropic pore architecture
- Author
-
Serena M. Best, Ruth E. Cameron, Christine J. Watson, Natalia Davidenko, Carlos F. Schuster, Thomas Gibb, Jonathan Campbell, Best, Serena [0000-0001-7866-8607], Watson, Christine [0000-0002-8548-5902], Cameron, Ruth [0000-0003-1573-4923], and Apollo - University of Cambridge Repository
- Subjects
Work (thermodynamics) ,Scaffold ,Materials science ,Scanning electron microscope ,Biomedical Engineering ,Nanotechnology ,Biochemistry ,law.invention ,Biomaterials ,Tissue engineering ,Biomimetic Materials ,law ,Phase (matter) ,Freezing ,oriented pore structure ,Animals ,Crystallization ,Composite material ,Anisotropy ,Molecular Biology ,Tissue Scaffolds ,General Medicine ,Microstructure ,tissue engineering ,scaffolds ,freeze-drying ,Microscopy, Electron, Scanning ,Cattle ,Collagen ,Porosity ,Biotechnology - Abstract
Sponge-like matrices with a specific three-dimensional structural design resembling the actual extracellular matrix of a particular tissue show significant potential for the regeneration and repair of a broad range of damaged anisotropic tissues. The manipulation of the structure of collagen scaffolds using a freeze-drying technique was explored in this work as an intrinsically biocompatible way of tailoring the inner architecture of the scaffold. The research focused on the influence of temperature gradients, imposed during the phase of crystallisation of collagen suspensions, upon the degree of anisotropy in the microstructures of the scaffolds produced. Moulding technology was employed to achieve differences in heat transfer rates during the freezing processes. For this purpose various moulds with different configurations were developed with a view to producing uniaxial and multi-directional temperature gradients across the sample during this process. Scanning electron microscopy analysis of different cross-sections (longitudinal and horizontal) of scaffolds revealed that highly aligned matrices with axially directed pore architectures were obtained where single unidirectional temperature gradients were induced. Altering the freezing conditions by the introduction of multiple temperature gradients allowed collagen scaffolds to be produced with complex pore orientations, and anisotropy in pore size and alignment.
- Published
- 2012
31. Conditional deletion of Stat3 in mammary epithelium impairs the acute phase response and modulates immune cell numbers during post‐lactational regression
- Author
-
Katherine Hughes, Christine J. Watson, Judith E. Allen, and Julie A Wickenden
- Subjects
mammary gland ,Mammary gland ,Cell Count ,Mice ,0302 clinical medicine ,Mast Cells ,signalling ,Mice, Knockout ,0303 health sciences ,Cell Death ,Mast cell ,Original Papers ,Up-Regulation ,medicine.anatomical_structure ,Mammary Epithelium ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,STAT3 Transcription Factor ,Inflammation ,macrophage ,Weaning ,Biology ,Cell Line ,Pathology and Forensic Medicine ,03 medical and health sciences ,breast cancer ,Mammary Glands, Animal ,Immune system ,involution ,medicine ,Animals ,Lactation ,Involution (medicine) ,acute-phase response ,Acute-Phase Reaction ,030304 developmental biology ,Wound Healing ,Innate immune system ,Stat3 ,Macrophages ,Epithelial Cells ,Macrophage Activation ,Immunity, Innate ,inflammation ,Immunology ,Cancer research ,Wound healing ,Gene Deletion - Abstract
Mammary gland regression following weaning (involution) is associated with extensive cell death and the acquisition of an inflammatory signature. Characterising the interplay between mammary epithelial cells, the re-emerging stroma, and immune cells has implications for the understanding of the pathogenesis of pregnancy associated breast cancer. Stat3 has a role in orchestrating cell death and involution, and we sought to determine whether expression of Stat3 by the mammary epithelium influences also the innate immune environment and inflammatory cell influx in the gland. We examined mice in which Stat3 is conditionally deleted only in the mammary epithelium. Distinct sets of genes associated with the acute phase response and innate immunity are markedly up-regulated during first phase involution in a Stat3-dependent manner. During second phase involution, chitinase 3-like 1, which has been associated with wound healing and chronic inflammatory conditions, is dramatically up-regulated by Stat3. Also at this time, the number of mammary macrophages and mast cells increases per unit area, and this increase is impaired in the absence of epithelial Stat3. Furthermore, expression of arginase-1 and Ym1, markers of alternatively activated macrophages, is significantly decreased in the absence of Stat3, whilst iNOS, a marker associated with classically activated macrophages, shows significantly increased expression in the Stat3 deleted glands. Thus, Stat3 is a key transcriptional regulator of genes associated with innate immunity and wound healing and influences mammary macrophage and mast cell numbers. The presence of epithelial Stat3 appears to polarize the macrophages and epithelial cells towards an alternatively activated phenotype, since in the absence of Stat3, the gland retains a phenotype associated with classically activated macrophages. These findings have relevance to the study of pregnancy associated breast cancer and the role of Stat3 signalling in recruitment of alternatively activated tumour-associated macrophages in breast cancer. Copyright © 2011 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2012
32. Cytokine signalling in mammary gland development
- Author
-
Christine J. Watson, Carrie H. Oliver, and Walid T. Khaled
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Mammary gland ,Biology ,Alveolar cells ,Mammary Glands, Animal ,Pregnancy ,Internal medicine ,Lactation ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mammary Glands, Human ,Th1-Th2 Balance ,Obstetrics and Gynecology ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Embryonic stem cell ,Epithelium ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,Cytokine ,Gene Expression Regulation ,Reproductive Medicine ,Cytokines ,Female ,Signal Transduction ,Transcription Factors ,Hormone - Abstract
Mammary gland development occurs in three distinct stages during the lifetime of the female mammal: in embryonic, pubertal and reproductive life. At each of these developmental stages, different signalling molecules induce changes in both the epithelium and the surrounding stroma. However, it is during pregnancy that the most dramatic changes occur, resulting in a massive increase in the number of epithelial cells and in their function. Pregnancy initiates the development of a new epithelial lineage, the alveolar cells, which form the milk-producing lobuloalveolar structures. These cells become redundant at the end of lactation and are removed in an exquisitely controlled process of tissue remodelling coupled with extensive cell death. All of these events require not only steroid hormones but also sequential signalling by cytokines. A recent surprising discovery was that the signalling pathways and cytokines that regulate lineage determination in T helper cells are also involved in mammary gland development during pregnancy.
- Published
- 2011
33. Stat3 controls lysosomal-mediated cell death in vivo
- Author
-
Anna Staniszewska, Nader Omidvar, Wenjing Li, James Turkson, Richard W. E. Clarkson, Richard A. Flavell, Valeria Poli, Peter A. Kreuzaler, Blandine Kedjouar, and Christine J. Watson
- Subjects
STAT3 Transcription Factor ,Programmed cell death ,Cell Membrane Permeability ,Cathepsin L ,Cell ,Mammary Neoplasms, Animal ,Cathepsin B ,STAT3 ,Mice ,Neoplasms ,involution ,medicine ,Animals ,lysosomal membrane permeabilization ,Involution (medicine) ,programmed cell death ,Crosses, Genetic ,Caspase ,Cathepsin ,Cell Death ,biology ,Cell Biology ,Cathepsins ,Immunohistochemistry ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Gene Expression Regulation ,Apoptosis ,biology.protein ,Female ,Lysosomes - Abstract
It is well established that lysosomes play an active role during the execution of cell death1. A range of stimuli can lead to lysosomal membrane permeabilization (LMP), thus inducing programmed cell death without involvement of the classical apoptotic programme2, 3. However, these lysosomal pathways of cell death have mostly been described in vitro or under pathological conditions4, 5, 6, 7. Here we show that the physiological process of post-lactational regression of the mammary gland is accomplished through a non-classical, lysosomal-mediated pathway of cell death. We found that, during involution, lysosomes in the mammary epithelium undergo widespread LMP. Furthermore, although cell death through LMP is independent of executioner caspases 3, 6 and 7, it requires Stat3, which upregulates the expression of lysosomal proteases cathepsin B and L, while downregulating their endogenous inhibitor Spi2A (ref. 8). Our findings report a previously unknown, Stat3-regulated lysosomal-mediated pathway of cell death under physiological circumstances. We anticipate that these findings will be of major importance in the design of treatments for cancers such as breast, colon and liver, where cathepsins and Stat3 are commonly overexpressed and/or hyperactivated respectively1, 9, 10.
- Published
- 2011
34. Stem cell marker prominin-1 regulates branching morphogenesis, but not regenerative capacity, in the mammary gland
- Author
-
Corinne A. Boulanger, Christine J. Watson, Lisa H. Anderson, Gilbert H. Smith, and Peter Carmeliet
- Subjects
Mammary gland ,Morphogenesis ,Mice, Nude ,Biology ,Stem cell marker ,Article ,Mice ,Mammary Glands, Animal ,Antigens, CD ,medicine ,Animals ,Regeneration ,AC133 Antigen ,Glycoproteins ,Stem Cells ,Prolactin receptor ,Flow Cytometry ,Mice, Mutant Strains ,Cell biology ,Neuroepithelial cell ,medicine.anatomical_structure ,Mammary Epithelium ,Immunology ,Female ,Stem cell ,Peptides ,Developmental Biology ,Adult stem cell - Abstract
Prominin-1 (Prom1) is recognized as a stem cell marker in several tissues, including blood, neuroepithelium, and gut, and in human and mouse embryos and many cancers. Although Prom1 is routinely used as a marker for isolating stem cells, its biological function remains unclear. Here we use a knockout model to investigate the role of Prom1 in the mammary gland. We demonstrate that complete loss of Prom1 does not affect the regenerative capacity of the mammary epithelium. Surprisingly, we also show that in the absence of Prom1, mammary glands have reduced ductal branching, and an increased ratio of luminal to basal cells. The effects of Prom1 loss in the mammary gland are associated with decreased expression of prolactin receptor and matrix metalloproteinase-3. These experiments reveal a novel, functional role for Prom1 that is not related to stem cell activity, and demonstrate the importance of tissue-specific characterization of putative stem cell markers.
- Published
- 2011
35. Remodeling mechanisms of the mammary gland during involution
- Author
-
Christine J. Watson and Peter A. Kreuzaler
- Subjects
Embryology ,medicine.medical_specialty ,Programmed cell death ,Mammary gland ,Gene Expression ,Inflammation ,Biology ,Models, Biological ,Fat pad ,Extracellular matrix ,Mammary Glands, Animal ,Pregnancy ,Internal medicine ,medicine ,Animals ,Humans ,Lactation ,Involution (medicine) ,Breast ,Innate immune system ,Cell Death ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Adipogenesis ,Female ,medicine.symptom ,Signal Transduction ,Developmental Biology - Abstract
The process of post-lactational regression, or involution, of the mammary gland is a complex event characterised by extensive death of the secretory epithelium coupled with remodelling of the extracellular matrix and adipogenesis to regenerate the fat pad. Associated with these events is an inflammatory cascade and acute phase response. The critical signalling pathways that regulated involution have been defined and a wide variety of genes have been shown to modulate the various processes involved, including cell death, phagocytosis, tissue remodelling and innate immune response.
- Published
- 2011
36. Collagen–hyaluronic acid scaffolds for adipose tissue engineering
- Author
-
Ruth E. Cameron, Jonathan Campbell, Eng San Thian, Natalia Davidenko, and Christine J. Watson
- Subjects
Scaffold ,Materials science ,Stromal cell ,Biomedical Engineering ,Adipose tissue ,Biocompatible Materials ,Biochemistry ,Biomaterials ,Mice ,Freeze-drying ,chemistry.chemical_compound ,3T3-L1 Cells ,Materials Testing ,Hyaluronic acid ,medicine ,Animals ,Humans ,Hyaluronic Acid ,Mammary Glands, Human ,Molecular Biology ,Cells, Cultured ,Adipogenesis ,Tissue Engineering ,Tissue Scaffolds ,Water ,General Medicine ,In vitro ,Extracellular Matrix ,Adipose Tissue ,chemistry ,Collagen ,Swelling ,medicine.symptom ,Porosity ,Biomarkers ,Biotechnology ,Biomedical engineering - Abstract
Three-dimensional (3-D) in vitro models of the mammary gland require a scaffold matrix that supports the development of adipose stroma within a robust freely permeable matrix. 3-D porous collagen-hyaluronic acid (HA: 7.5% and 15%) scaffolds were produced by controlled freeze-drying technique and crosslinking with 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride. All scaffolds displayed uniform, interconnected pore structure (total porosity approximately 85%). Physical and chemical analysis showed no signs of collagen denaturation during the formation process. The values of thermal characteristics indicated that crosslinking occurred and that its efficiency was enhanced by the presence of HA. Although the crosslinking reduced the swelling of the strut material in water, the collagen-HA matrix as a whole tended to swell more and show higher dissolution resistance than pure collagen samples. The compressive modulus and elastic collapse stress were higher for collagen-HA composites. All the scaffolds were shown to support the proliferation and differentiation 3T3-L1 preadipocytes while collagen-HA samples maintained a significantly increased proportion of cycling cells (Ki-67+). Furthermore, collagen-HA composites displayed significantly raised Adipsin gene expression with adipogenic culture supplementation for 8 days vs. control conditions. These results indicate that collagen-HA scaffolds may offer robust, freely permeable 3-D matrices that enhance mammary stromal tissue development in vitro.
- Published
- 2010
37. Constitutively active Stat3 enhances neu-mediated migration and metastasis in mammary tumors via upregulation of Cten
- Author
-
Diego Maritano, Marco Demaria, Sara Pensa, Federica Cavallo, Tania Pannellini, Paolo Provero, Elena Quaglino, Alessandra Voster, Christine J. Watson, James Turkson, Valeria Poli, Isaia Barbieri, Piero Musiani, Damage and Repair in Cancer Development and Cancer Treatment (DARE), and Restoring Organ Function by Means of Regenerative Medicine (REGENERATE)
- Subjects
Male ,STAT3 Transcription Factor ,Cancer Research ,Lung Neoplasms ,Transcription, Genetic ,Receptor, ErbB-2 ,Mice, Transgenic ,Breast Neoplasms ,HER2/Neu ,HER2/neu ,Transgenic ,Proinflammatory cytokine ,Cell Line ,Experimental ,Mice ,breast cancer ,ErbB-2 ,Downregulation and upregulation ,Genetic ,Cell Movement ,Cell Line, Tumor ,Tensins ,Stat3 ,C-Terminal Tensin-like ,Tensin ,Animals ,Humans ,Phosphorylation ,STAT3 ,Cell Nucleus ,Tumor ,biology ,Mouse mammary tumor virus ,Mammary Neoplasms ,Microfilament Proteins ,Mammary Neoplasms, Experimental ,Cell migration ,biology.organism_classification ,Up-Regulation ,Oncology ,biology.protein ,Cancer research ,STAT protein ,Female ,Transcription ,Receptor - Abstract
The transcription factor signal transducer and activator of transcription 3 (STAT3) is constitutively activated in tumors of different origin, but the molecular bases for STAT3 requirement are only partly understood. To evaluate the contribution of enhanced Stat3 activation in a controlled model system, we generated knock-in mice wherein a mutant constitutively active Stat3C allele replaces the endogenous wild-type allele. Stat3C could enhance the tumorigenic power of the rat Neu oncogene in mouse mammary tumor virus (MMTV)–Neu transgenic mice, triggering the production of earlier onset, more invasive mammary tumors. Tumor-derived cell lines displayed higher migration, invasion, and metastatic ability and showed disrupted distribution of cell-cell junction markers mediated by Stat3-dependent overexpression of the COOH terminal tensin-like (Cten) focal adhesion protein, which was also significantly upregulated in Stat3C mammary tumors. Importantly, the proinflammatory cytokine interleukin-6 could mediate Cten induction in MCF10 cells in an exquisitely Stat3-dependent way, showing that Cten upregulation is a feature of inflammation-activated Stat3. In light of the emerging pivotal role of Stat3 in connecting inflammation and cancer, our identification of Cten as a Stat3-dependent mediator of migration provides important new insights into the oncogenic role of Stat3, particularly in the breast. Cancer Res; 70(6); 2558–67
- Published
- 2010
38. PML: A tumor suppressor that regulates cell fate in mammary gland
- Author
-
Christine J. Watson, Tina Rich, and Wenjing Li
- Subjects
Population ,Estrogen receptor ,Promyelocytic Leukemia Protein ,Biology ,Cell fate determination ,stat ,law.invention ,Mice ,Mammary Glands, Animal ,law ,Animals ,Protein Isoforms ,Progenitor cell ,education ,Molecular Biology ,Progenitor ,education.field_of_study ,Tumor Suppressor Proteins ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Cell biology ,STAT Transcription Factors ,Immunology ,Suppressor ,Female ,Stem cell ,Transcription Factors ,Developmental Biology - Abstract
The tumor suppressor promyelocytic leukemia protein (PML) is possibly unique in that its tumor suppressive functions may be attributed to both the protein and the conspicuous nuclear bodies (PML-NBs) that PML builds. Untangling the role of either the protein or its domain in cell fate has been a decade long task which has just received new impetus from developmental biologists. PML appears to play a central role in regulating stem and progenitor cell fate in tissues as diverse as blood, brain and breast. Our studies have uncovered an inverse relationship between the activity of certain Stat transcription factors and PML in controlling normal mammary gland development and the regulation of lineage commitment. Genetic loss of Pml delays differentiation of the milk-producing alveolar cells and disrupts ductal development, defects which may result from a skewing of the progenitor population to favor estrogen receptor positive cells (ERalpha). This is of considerable interest as ERalpha cells are non-cycling in normal breast while promiscuous cell cycle entry is a feature of these cells in breast cancer. These data begin to show the cell types and tissues that are most sensitive to PML dose and provide new perspectives for the regulation of mammary gland development and tumorigenesis.
- Published
- 2009
39. The Role of Cathepsins in Involution and Breast Cancer
- Author
-
Christine J. Watson and Peter A. Kreuzaler
- Subjects
Cancer Research ,medicine.medical_specialty ,Breast Neoplasms ,Endosomes ,Biology ,medicine.disease_cause ,Cathepsin B ,Metastasis ,Mice ,Mammary Glands, Animal ,Pregnancy ,Internal medicine ,Lysosome ,medicine ,Animals ,Humans ,Lactation ,Involution (medicine) ,Breast ,Neoplasm Metastasis ,Serpins ,Mice, Knockout ,Cathepsin ,Cell Death ,Proteolytic enzymes ,Mammary Neoplasms, Experimental ,Cancer ,medicine.disease ,Cathepsins ,Cystatins ,Neoplasm Proteins ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Cancer research ,Female ,Lysosomes ,Carcinogenesis - Abstract
Cysteine cathepsins are proteolytic enzymes that reside in endolysosomal vesicles. Some are expressed constitutively while others are transcriptionally regulated. However, the expression and subcellular localization of cathepsins changes during cancer progression and cathepsins have been shown to be causally involved in various aspects of tumorigenesis including metastasis. The use of mouse models of breast cancer genetically ablated for cathepsin B has shown that both the growth of the primary tumor and the extend of lung metastasis is reduced by the loss of cathepsin B. The role of cathepsins in involution of the mammary gland has received little attention although it is clear that cathepsins are involved in tissue remodeling in the second phase of involution. We discuss here the roles of cathepsins and their endogenous inhibitors in breast tumorigenesis and post-lactational involution.
- Published
- 2009
40. Three-dimensional culture models of mammary gland
- Author
-
Jonathan Campbell and Christine J. Watson
- Subjects
Basement membrane ,Transplantation ,Embryology ,medicine.medical_specialty ,Mammary gland ,Biomedical Engineering ,Myoepithelial cell ,Review ,Biology ,Epithelium ,Cell biology ,Extracellular matrix ,medicine.anatomical_structure ,Endocrinology ,Tissue engineering ,Stroma ,Cell culture ,Internal medicine ,medicine ,Developmental Biology - Abstract
The mammary gland is a complex tissue comprised of a branching network of ducts embedded within an adipocyte-rich stroma. The ductal epithelium is a bi-layer of luminal and myoepithelial cells, the latter being in contact with a basement membrane. During pregnancy, tertiary branching occurs and lobuloalveolar structures, which produce milk during lactation, form in response to hormonal and cytokine signals. Postlactational regression is characterized by extensive cell death and tissue remodeling. These complex developmental events have been difficult to mimic in cell culture although many useful culture models exist. Recently, considerable advances in three-dimensional modelling of the mammary gland have been made with the use of collagen and other biomaterials for the study of branching morphogenesis and tumorigenesis, techniques which may enable rapid advances in our understanding of both basic biology and the study of cancer therapeutics.
- Published
- 2009
41. Mammary development in the embryo and adult: a journey of morphogenesis and commitment
- Author
-
Christine J. Watson and Walid T. Khaled
- Subjects
T-Lymphocytes ,Mesenchyme ,Mammary gland ,Morphogenesis ,Biology ,Mice ,Mammary Glands, Animal ,Stroma ,Pregnancy ,medicine ,Animals ,Sexual Maturation ,Progenitor cell ,Molecular Biology ,Body Patterning ,Reproduction ,Stem Cells ,Epithelial Cells ,Embryo ,Embryonic stem cell ,Epithelium ,Cell biology ,medicine.anatomical_structure ,Immunology ,Female ,Signal Transduction ,Developmental Biology - Abstract
Mammary gland development occurs through distinctive stages throughout embryonic and pubertal development and reproductive life. At each stage,different signals are required to induce changes in both the epithelium and the surrounding mesenchyme/stroma. Recent studies have provided new insights into the origin, specification and fate of mammary stem and progenitor cells and into how the differentiated lineages that comprise the functional mammary gland are determined. The development of new tools and culture techniques has also enabled the factors that influence branching morphogenesis in the embryonic and pubertal gland to be identified. A surprising recent discovery has been that mammary epithelial cells commit to differentiated lineages using the same signalling pathways that regulate lineage determination in T helper cells.
- Published
- 2008
42. The IL-4/IL-13/Stat6 signalling pathway promotes luminal mammary epithelial cell development
- Author
-
Fiona O. Baxter, Andrew N. J. McKenzie, Eliot Read, Amelia J. Brennan, Paul J. Came, Sandra E. Nicholson, Walid T. Khaled, Naomi S. Sprigg, and Christine J. Watson
- Subjects
medicine.medical_treatment ,Morphogenesis ,Mice, SCID ,Biology ,Models, Biological ,Mice ,Mammary Glands, Animal ,Immune system ,Downregulation and upregulation ,Mice, Inbred NOD ,Pregnancy ,medicine ,Animals ,Interferon gamma ,Molecular Biology ,Cells, Cultured ,Interleukin 4 ,Mice, Knockout ,Mice, Inbred BALB C ,Interleukin-13 ,Epithelial Cells ,Cell biology ,Mice, Inbred C57BL ,Cytokine ,Interleukin 13 ,Interleukin 12 ,Female ,Interleukin-4 ,STAT6 Transcription Factor ,Signal Transduction ,Developmental Biology ,medicine.drug - Abstract
Naïve T helper cells differentiate into Th1 and Th2 subsets, which have unique cytokine signatures, activators and transcriptional targets. The Th1/Th2 cytokine milieu is a key paradigm in lineage commitment, and IL-4(Il4), IL-13 (Il13) and Stat6 are important mediators of Th2 development. We show here, for the first time, that this paradigm applies also to mammary epithelial cells, which undergo a switch from Th1 to Th2 cytokine production upon the induction of differentiation. Thus, the Th1 cytokines IL-12 (Il12),interferon gamma (INFγ; also known as Ifng) and Tnfα are downregulated concomitantly with the upregulation of the Th2 cytokines IL-4,IL-13 and IL-5 (Il5) as epithelial cells commit to the luminal lineage. Moreover, we show that Th2 cytokines play a crucial role in mammary gland development in vivo, because differentiation and alveolar morphogenesis are reduced in both Stat6 and IL-4/IL-13 doubly deficient mice during pregnancy. This unexpected discovery demonstrates a role for immune cell cytokines in epithelial cell fate and function, and adds an unexpected tier of complexity to the previously held paradigm that steroid and peptide hormones are the primary regulators of mammary gland development.
- Published
- 2007
43. BCL11A is a triple-negative breast cancer gene with critical functions in stem and progenitor cells
- Author
-
H. Raza Ali, Andy Futreal, Oscar M. Rueda, Pentao Liu, Neal G. Copeland, John Stingl, Xiongfeng Chen, Juexuan Wang, Michael R. Stratton, Carlos Caldas, Christine J. Watson, Walid T. Khaled, Nancy A. Jenkins, Fazal Hadi, Samuel Aparicio, Suet-Feung Chin, Song Choon Lee, Yong Yu, Khaled, Walid [0000-0001-9068-5776], Rueda Palacio, Oscar [0000-0003-0008-4884], Hadi, Fazal [0000-0003-3553-817X], Chin, Suet-Feung [0000-0001-5697-1082], Watson, Christine [0000-0002-8548-5902], Caldas, Carlos [0000-0003-3547-1489], and Apollo - University of Cambridge Repository
- Subjects
Poor prognosis ,Cell Survival ,9,10-Dimethyl-1,2-benzanthracene ,General Physics and Astronomy ,Triple Negative Breast Neoplasms ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,Mammary Glands, Animal ,0302 clinical medicine ,Cell Line, Tumor ,Animals ,Humans ,Progenitor cell ,Gene ,Triple-negative breast cancer ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Stem Cells ,Nuclear Proteins ,General Chemistry ,Prognosis ,Immunohistochemistry ,3. Good health ,DNA-Binding Proteins ,Repressor Proteins ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Epidemiologic data ,Stem cell ,Carrier Proteins ,Neoplasm Transplantation - Abstract
Triple-negative breast cancer (TNBC) has poor prognostic outcome compared with other types of breast cancer. The molecular and cellular mechanisms underlying TNBC pathology are not fully understood. Here, we report that the transcription factor BCL11A is overexpressed in TNBC including basal-like breast cancer (BLBC) and that its genomic locus is amplified in up to 38% of BLBC tumours. Exogenous BCL11A overexpression promotes tumour formation, whereas its knockdown in TNBC cell lines suppresses their tumourigenic potential in xenograft models. In the DMBA-induced tumour model, Bcl11a deletion substantially decreases tumour formation, even in p53-null cells and inactivation of Bcl11a in established tumours causes their regression. At the cellular level, Bcl11a deletion causes a reduction in the number of mammary epithelial stem and progenitor cells. Thus, BCL11A has an important role in TNBC and normal mammary epithelial cells. This study highlights the importance of further investigation of BCL11A in TNBC-targeted therapies., Triple-negative breast cancers (TNBCs) tend to have poor prognosis; however, the mechanisms underlying TNBC pathology are not well understood. Here the authors utilize epidemiologic data and animal models to demonstrate an important role for BCL11A in the genesis and propagation of TNBCs.
- Published
- 2015
44. IKKβ/2 induces TWEAK and apoptosis in mammary epithelial cells
- Author
-
Marion Huth, Fiona O. Baxter, Kathrine Abell, Paul J. Came, Blandine Kedjouar, Manolis Pasparakis, Christine J. Watson, and Klaus Rajewsky
- Subjects
Apoptosis ,Caspase 3 ,Biology ,Ligands ,Mice ,Mammary Glands, Animal ,Animals ,Involution (medicine) ,Promoter Regions, Genetic ,Molecular Biology ,Protein kinase B ,Transcription factor ,Cytokine TWEAK ,Regulation of gene expression ,Gene Expression Regulation, Developmental ,Epithelial Cells ,I-kappa B Kinase ,Tumor Necrosis Factors ,Cancer research ,Female ,Signal transduction ,Apoptosis Regulatory Proteins ,Gene Deletion ,Signal Transduction ,Developmental Biology - Abstract
The Nuclear Factor-kappaB (NF-kappaB) family of transcription factors are ubiquitously expressed and control a wide range of cellular responses, including apoptosis, proliferation, differentiation, inflammation and immunity. Here, we investigated the function of the NF-kappaB upstream regulator IkappaB kinase 2/beta (IKK2) in apoptosis regulation in the normal physiological setting of regressing mammary gland. Conditional deletion of the gene encoding IKK2 resulted, surprisingly, in delayed apoptosis and remodelling, and abrogation of caspase 3 cleavage. This failure to induce involution was associated with reduced expression, within 24 hours of involution, of the death receptor (DR) ligand TNF and its receptor TNFR1, which are known NF-kappaB targets. This was associated with elevated levels of active AKT and phosphorylated FOXO3a. Furthermore, we show that expression of TWEAK, another DR ligand, is dramatically downregulated, even in heterozygous IKK2 mammary glands. Unlike other DR ligands, the TWEAK promoter has six consensus FOXO-binding sites, further suggesting that it is differentially regulated. Interestingly, a cleaved form of TWEAK is upregulated during involution. This unexpected function of the IKK2/NF-kappaB pathway as a regulator of TWEAK expression and inducer of apoptosis has significant consequences for future therapeutic approaches for cancer and inflammatory diseases.
- Published
- 2006
45. The Genes Induced by Signal Transducer and Activators of Transcription (STAT)3 and STAT5 in Mammary Epithelial Cells Define the Roles of these STATs in Mammary Development
- Author
-
Marion P. Boland, Christine J. Watson, Tom C. Freeman, Paul G. Tiffen, Ekaterini A. Kritikou, Richard W. E. Clarkson, and Jennifer M. Lee
- Subjects
STAT3 Transcription Factor ,Programmed cell death ,Recombinant Fusion Proteins ,Cellular differentiation ,Apoptosis ,Leukemia Inhibitory Factor ,Mice ,Mammary Glands, Animal ,Endocrinology ,STAT5 Transcription Factor ,Animals ,STAT3 ,Molecular Biology ,Transcription factor ,Cells, Cultured ,STAT5 ,Oligonucleotide Array Sequence Analysis ,Cell Death ,biology ,Ccaat-enhancer-binding proteins ,Interleukin-6 ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Epithelial Cells ,General Medicine ,DNA Gyrase ,biology.protein ,Cancer research ,Female ,Dimerization ,Leukemia inhibitory factor - Abstract
Prolactin and leukemia inhibitory factor (LIF) have different roles in the adult mammary gland, which are mediated in part by the signal transducers and activators of transcription (STAT)5 and STAT3. In vivo studies have shown that STAT5 contributes to prolactin-dependent lobuloalveolar development and lactation whereas STAT3 mediates LIF-dependent epithelial apoptosis during postlactational involution. To understand the molecular basis of these STAT-dependent pathways, we demonstrate the ligand-independent effects of STAT5 and STAT3 in mammary epithelial cells in vitro and also identify the genes regulated by these related transcription factors. Thus, using conditionally active STAT3- or STAT5a-GyraseB fusion proteins, we observed that enforced and specific dimerization of STAT3 induced apoptosis whereas STAT5 induced differentiation of mammary epithelial cells. Furthermore, STAT5 attenuated apoptosis mediated by LIF, the physiological inducer of STAT3. Microarray analysis of STAT3- and STAT5-induced genes using this system demonstrated a marked specificity, which reflected their different physiological effects in vitro and in vivo. STAT5-specific gene targets included the milk protein genes alpha-casein and kallikrein-8 and the survival factors prosaposin and Grb10. STAT3-specific genes included the apoptosis regulators CCAAT enhancer binding protein-delta, phosphatidylinositol 3-kinase-regulatory subunits, purine nucleoside phosphorylase, and c-fos. These data illustrate that specific activation of STAT3 and STAT5 alone is sufficient to induce and suppress apoptosis, respectively, and that these transcription factors elicit their actions by inducing distinct subsets of target genes in mammary epithelial cells.
- Published
- 2006
46. A dual, non-redundant, role for LIF as a regulator of development and STAT3-mediated cell death in mammary gland
- Author
-
Andrew M. Sharkey, Kathrine Abell, Paul J. Came, Elizabeth Anderson, Ekaterini A. Kritikou, Christine J. Watson, and Richard W. E. Clarkson
- Subjects
STAT3 Transcription Factor ,endocrine system ,Programmed cell death ,Leukemia Inhibitory Factor Receptor alpha Subunit ,Receptors, OSM-LIF ,Mammary gland ,MAP Kinase Kinase 1 ,Apoptosis ,Protein Serine-Threonine Kinases ,Biology ,Leukemia Inhibitory Factor ,Mice ,Mammary Glands, Animal ,Downregulation and upregulation ,Contactins ,Nitriles ,Butadienes ,STAT5 Transcription Factor ,medicine ,Animals ,Involution (medicine) ,Receptors, Cytokine ,STAT3 ,Neural Cell Adhesion Molecules ,Molecular Biology ,reproductive and urinary physiology ,Mitogen-Activated Protein Kinase Kinases ,Lymphokines ,Interleukin-6 ,urogenital system ,Activator (genetics) ,Caseins ,Gene Expression Regulation, Developmental ,Milk Proteins ,Growth Inhibitors ,Cell biology ,DNA-Binding Proteins ,STAT1 Transcription Factor ,medicine.anatomical_structure ,embryonic structures ,Trans-Activators ,biology.protein ,Cancer research ,Phosphorylation ,Female ,Receptors, Progesterone ,hormones, hormone substitutes, and hormone antagonists ,Developmental Biology - Abstract
STAT3 is the key mediator of apoptosis in mammary gland. We demonstrate here that LIF is the physiological activator of STAT3, because in involuting mammary glands of Lif -;/-; mice, pSTAT3 is absent and the STAT3 target, C/EBPδ, is not upregulated. Similar to Stat3 knockouts, Lif -;/-; mammary glands exhibit delayed involution, reduced apoptosis and elevated levels of p53. Significantly, Lif -;/-; glands display precocious development during pregnancy, when pSTAT3 is not normally detected. We show that pERK1/2 is significantly reduced in Lif -;/-; glands at this time, suggesting that at this stage LIF mediates its effects through pERK1/2. Inhibition of LIF-mediated ERK1/2 phosphorylation potentiates the proapoptotic effects of STAT3. LIF therefore signals alternately through ERK1/2, then STAT3, to regulate mammary growth and apoptosis.
- Published
- 2003
47. A 3-D in vitro co-culture model of mammary gland involution
- Author
-
Jonathan Campbell, Natalia Davidenko, Laur-Alexandru Botos, Ruth E. Cameron, Christine J. Watson, Timothy J. Sargeant, Cameron, Ruth [0000-0003-1573-4923], Watson, Christine [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
STAT3 Transcription Factor ,medicine.medical_specialty ,Programmed cell death ,Stromal cell ,medicine.medical_treatment ,Mammary gland ,Biophysics ,Oncostatin M ,In Vitro Techniques ,Biochemistry ,Mice ,Mammary Glands, Animal ,Internal medicine ,3T3-L1 Cells ,medicine ,Animals ,Involution (medicine) ,Mammary gland involution ,Microscopy, Confocal ,Cell Death ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Regeneration (biology) ,Macrophages ,Epithelial Cells ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Cytokine ,Cell culture ,Female ,Stromal Cells - Abstract
Involution is a process whereby the mammary gland undergoes extensive tissue remodelling involving exquisitely coordinated cell death, extracellular matrix degradation and adipose tissue regeneration following the weaning of offspring. These processes are mediated in part through Jak/Stat signalling pathways, which can be deregulated in breast cancer. Synthetic in vitro analogues of the breast could become important tools for studying tumorigenic processes, or as personalized drug discovery platforms and predictors of therapeutic response. Ideally, such models should support 3D neo-tissue formation, so as to recapitulate physiological organ function, and be compatible with high-throughput screening methodologies. We have combined cell lines of epithelial, stromal and immunological origin within engineered porous collagen/hyaluronic acid matrices, demonstrating 3D-specific molecular signatures. Furthermore seeded cells form mammary-like branched tissues, with lobuloalveolar structures that undergo inducible involution phenotypes reminiscent of the native gland under hormonal/cytokine regulation. We confirm that autophagy is mediated within differentiated mammary epithelial cells in a Stat-dependent manner at early time points following the removal of a prolactin stimulus (H/WD). In addition, epithelial cells express markers of an M2 macrophage lineage under H/WD, a process that is attenuated with the introduction of the monocyte/macrophage cell line RAW 264.7. Thus, such 3D models are suitable platforms for studying cell-cell interactions and cell death mechanisms in relation to cancer.
- Published
- 2014
48. Engineering mammary gland in vitro models for cancer diagnostics and therapy
- Author
-
Christine J. Watson, Robert D. Hume, and Jonathan Campbell
- Subjects
Pathology ,medicine.medical_specialty ,Mammary gland ,Complex disease ,Pharmaceutical Science ,Breast Neoplasms ,Biology ,In Vitro Techniques ,Models, Biological ,Breast cancer ,Mammary Glands, Animal ,Drug Discovery ,medicine ,Animals ,Humans ,Mammary Glands, Human ,Tissue Engineering ,Cancer ,medicine.disease ,In vitro ,Stem cell niche ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Cancer research ,Molecular Medicine ,Female - Abstract
Breast cancer is a complex disease with many distinct subtypes being recognized on the basis of histological features and molecular signatures. It is difficult to predict how cancers will respond to therapy, which results in many women receiving unnecessary or inappropriate treatment. Advances in materials science and tissue engineering are leading the development of complex in vitro 3D breast tissue models that will increase our understanding of normal development and tumorigenic mechanisms. Ultimately, platforms that support primary tissue culture could readily be adapted to form high-throughput drug screening tools for personalized medicine. This review will summarize the control of mammary gland phenotype within in vitro 3D environments, in the context of a detailed analysis of mammary gland development and stem and progenitor cell controlled tumorigenesis.
- Published
- 2014
49. Stat3 controls cell death during mammary gland involution by regulating uptake of milk fat globules and lysosomal membrane permeabilization
- Author
-
Timothy J. Sargeant, Christine J. Watson, Bethan Lloyd-Lewis, Henrike K. Resemann, Antonio Ramos-Montoya, Jeremy N. Skepper, Watson, Christine [0000-0002-8548-5902], and Apollo - University of Cambridge Repository
- Subjects
STAT3 Transcription Factor ,Programmed cell death ,Apoptosis ,Mice, Transgenic ,Vacuole ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Mammary Glands, Animal ,Phagocytosis ,Lipid droplet ,Animals ,Involution (medicine) ,Secretion ,Mammary gland involution ,030304 developmental biology ,Glycoproteins ,Cathepsin ,Mice, Knockout ,0303 health sciences ,Cell Death ,Biological Transport ,Epithelial Cells ,Cell Biology ,Lipid Droplets ,Cathepsins ,Cell biology ,Mice, Inbred C57BL ,030220 oncology & carcinogenesis ,Female ,Glycolipids ,Lysosomes - Abstract
We have previously demonstrated that Stat3 regulates lysosomal-mediated programmed cell death (LM-PCD) during mouse mammary gland involution in vivo. However, the mechanism that controls the release of lysosomal cathepsins to initiate cell death in this context has not been elucidated. We show here that Stat3 regulates the formation of large lysosomal vacuoles that contain triglyceride. Furthermore, we demonstrate that milk fat globules (MFGs) are toxic to epithelial cells and that, when applied to purified lysosomes, the MFG hydrolysate oleic acid potently induces lysosomal leakiness. Additionally, uptake of secreted MFGs coated in butyrophilin 1A1 is diminished in Stat3-ablated mammary glands and loss of the phagocytosis bridging molecule MFG-E8 results in reduced leakage of cathepsins in vivo. We propose that Stat3 regulates LM-PCD in mouse mammary gland by switching cellular function from secretion to uptake of MFGs. Thereafter, perturbation of lysosomal vesicle membranes by high levels of free fatty acids results in controlled leakage of cathepsins culminating in cell death.
- Published
- 2014
50. Breast cancer: the menacing face of Janus kinase
- Author
-
Christine J. Watson and Katherine Hughes
- Subjects
medicine.medical_specialty ,Cell Survival ,Mammary Neoplasms, Animal ,Suppressor of Cytokine Signaling Proteins ,medicine.disease_cause ,Mice ,Breast cancer ,Suppressor of Cytokine Signaling 1 Protein ,Growth factor receptor ,Internal medicine ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Janus kinase 2 ,biology ,Prolactin receptor ,Estrogen Receptor alpha ,Cell Biology ,Janus Kinase 2 ,medicine.disease ,Endocrinology ,Editorial ,STAT1 Transcription Factor ,Cancer cell ,biology.protein ,Cancer research ,Neoplastic Stem Cells ,Female ,Carcinogenesis ,Estrogen receptor alpha ,Heterocyclic Compounds, 3-Ring ,Tamoxifen ,medicine.drug ,Signal Transduction - Abstract
Janus, the two-faced Roman god of gates and doors, beginnings and endings, symbolizes important transitions. How appropriate therefore that Janus kinase 2 has been discovered to control the transition from normal mammary gland development to breast cancer in a recent manuscript from the Schreiber laboratory published in Cell Death and Differentiation.1 The lifetime risk of breast cancer for women in the western world is 1 in 8 and the incidence rate is rising toward epidemic levels with 1.4 million women diagnosed annually with this disease worldwide http://www.cancerresearchuk.org/cancer-info/cancerstats/world/breast-cancer-world/. Breast cancer is a heterogeneous disease with three main histological subtypes: estrogen receptor alpha/progesterone receptor (ERα/PR) positive, HER2 overexpressing, and triple negative tumors, which do not express ERα, PR or HER2. Six subtypes have been identified on the basis of gene expression signatures, each with a different prognosis.2 Patients with luminal subtype tumors, which are ERα positive and account for over 70% of breast cancers, have the best prognosis and are treated clinically with anti-estrogen therapies such as tamoxifen or aromatase inhibitors. However, despite an overall good prognosis for ERα-positive breast cancer, ∼25% of women develop resistance to anti-estrogen therapy, have recurrent tumors and succumb to metastatic disease.3 Furthermore, ERα+ breast cancer is itself heterogeneous. There is thus a pressing need to understand the origins of ERα+ tumors and the molecular mechanisms that give rise to their diversity and differential response to anti-estrogen therapy. Murine breast cancer models have been invaluable in delineation of both the genes and signaling pathways that regulate tumorigenesis. However, although ERα+ tumors are the most common type of breast cancer in women, there is a paucity of experimental mouse models of ERα+ tumors. Recently, the Schreiber laboratory described a new mouse model of ERα+ mammary carcinoma that arises in mice deficient for Stat1.4 Stat1 is a member of the Stat family of latent transcription factors, which bind to cytokine and growth factor receptors on their engagement by ligand, resulting in activation of receptor-associated JAK kinases that in turn tyrosine phosphorylate Stats, which dimerize, translocate to the nucleus and bind to promoters of target genes. Genetic ablation of Stat1 resulted in the spontaneous development of mammary tumors, with a long latency, that recapitulate many of the features and gene expression profiles of ERα+ luminal breast cancer. Thus, Stat1 is an unexpected tumor suppressor in mammary gland and this correlates with reduced expression levels of STAT1 in 45% of ERα+ human breast tumors, suggesting that Stat1−/− mice may be a useful model of ERα+ breast cancer. Conversely, other members of the Stat family of transcription factors, in particular Stat3 and Stat5, are potent oncogenes in mammary gland and appear to have reciprocal effects on target gene expression.5, 6, 7 During normal mammary gland development, Stat5 is essential for alveologenesis, the process by which differentiated milk-producing cells arise during pregnancy,8 whereas Stat3 is a critical mediator of cell death during post-lactational regression of the gland.9, 10 Stats 3 and 5 are generally thought of as having opposing functions and, despite recognizing a similar DNA-binding motif, bind distinct promoters.10 This Stat1−/− ERα+ tumor model has now been characterized in more detail and the authors show that control of Jak2 activity by the Stat1-SOCS1 axis is essential to maintain mammary gland homeostasis and that deletion of Stat1 and the concomitant loss of SOCS1, a negative regulator of Jak2, results in hyperactivation of Jak2 and unopposed signaling through the prolactin receptor (PrlR). This corroborates a previous study showing that loss of a single allele of SOCS1 can rescue lactation failure that occurs in PrlR+/− mice.11, 12 Interestingly, persistent PrlR signaling is a feature also of human ERα+ and ERα− breast cancer cells13 and elevated serum levels of prolactin (Prl) have been associated with increased risk of invasive ERα+ tumors and poor long-term survival.14 Furthermore, over 95% of human breast cancers overexpress PrlR and human breast cancer cells have been shown to upregulate local synthesis of Prl.15 The primary transcription factor downstream of PrlR in mammary gland is Stat5a.16 However, in Stat1−/− tumors, pJak2, pStat3 and pStat5a/b were all detected with pStat3 and pStat5 being observed in a proportion of the tumor cells raising the possibility that they are either co-activated in a subset of cells or that they are activated in two discrete populations. This distinction may be critically important to understanding the perturbation of signaling pathways in these tumors. Previous studies demonstrated that breast tumors exhibiting both activated STAT3 and STAT5 were more differentiated than tumors with just pSTAT3.5 As only Stat5 engages with the PrlR in normal mammary gland during pregnancy, when Stat3 is present at high levels but is not phosphorylated, this suggests that active Jak2 is bound also to receptors that engage Stat3 such as gp130/LIFR in Stat1-deficient mammary cells. It is well established that both Stat1 and Stat3 are activated downstream of the common gp130 receptor chain but in many contexts, gp130 is preferentially bound by Stat3, so in this instance it is interesting that deficiency of Stat1 has a profound effect on levels of pStat3.17 Using knockdown or inhibition of Jak2, the authors demonstrated that Jak2 is required for phosphorylation of Stat3 and Stat5 and that persistent activation of the PrlR-Jak2-Stat3/5a/5b axis provides a potent survival signal to tumor-derived cells that have presumably become addicted to Stat3 and/or Stat5a/5b, as observed in other human tumors and cell lines.18 As these studies utilized cell lines, the requirement for PrlR signaling in vivo was investigated by treating Stat1−/− tumor-bearing animals with a Jak2 inhibitor. This blocked, as anticipated, pStat3 and pStat5 and resulted in an increase in cleaved caspase 3-positive cells and a remarkable and prolonged inhibition of tumor growth for 7 months following inhibitor withdrawal. Furthermore, Jak2 drives tumor growth in both hormone-dependent and hormone-independent Stat1−/− tumors. This interesting and important work provides a useful model for further studies on the initiation and progression of ERα+ breast tumors (Figure 1). JAK2 inhibitors are currently undergoing clinical trials and these could be a valuable addition to the clinician's armory for tamoxifen-resistant breast cancers. Also, given the demonstration in this manuscript of the prophylactic efficacy of a JAK2 inhibitor, women at risk for breast cancer could receive anti-JAK2 therapy. In this context, it is notable that Jak2 has been shown to be required for initiation but not the maintenance of mammary tumors driven by Prl overexpression,19 as genetic ablation of Jak2 before, but not after, neoplastic transformation abolished tumorigenesis. The discrepancy between these two studies suggests that signaling through Jak2, downstream of receptors other than PrlR in the Stat1−/− mice, results in tumors that are dependent on Jak2. Figure 1 Summary of outcomes downstream of the prolactin receptor and JAK2 in breast cancer and in normal mammary gland. During pregnancy, engagement of the prolactin receptor by its ligand prolactin results in phosphorylation of Stat5, which regulates alveologenesis ... Further work is required to determine whether inhibition or ablation of either Stat5 or Stat3 alone is sufficient to inhibit tumor growth or whether it is the combination of both of these Stats that is required to generate ERα+ tumors. Given the competition between Stat5 and Stat3 for binding to specific promoters, and their overall distinct sets of transcriptional targets, it will be informative to investigate whether relative levels of these Stats dictate the clinical outcome. This ERα+ breast tumor model described by the Schreiber laboratory further endorses the view that there is an intricate interplay between Janus kinases and their corresponding Stats during mammary tumorigenesis. The irrefutable conclusion that can be drawn from this work is that perturbing the delicate balance between the activities of JAK2, STAT1, STAT3 and STAT5 leads to breast cancer development and targeting this pathway provides opportunities for therapeutic intervention.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.