Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(8): 1708-1728, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36931265

ABSTRACT

Breast cancer remains a leading cause of cancer-related mortality in women, reflecting profound disease heterogeneity, metastasis, and therapeutic resistance. Over the last decade, genomic and transcriptomic data have been integrated on an unprecedented scale and revealed distinct cancer subtypes, critical molecular drivers, clonal evolutionary trajectories, and prognostic signatures. Furthermore, multi-dimensional integration of high-resolution single-cell and spatial technologies has highlighted the importance of the entire breast cancer ecosystem and the presence of distinct cellular "neighborhoods." Clinically, a plethora of new targeted therapies has emerged, now being rapidly incorporated into routine care. Resistance to therapy, however, remains a crucial challenge for the field.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Gene Expression Profiling , Genomics , Transcriptome , Drug Resistance, Neoplasm
2.
Physiol Rev ; 100(2): 489-523, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31539305

ABSTRACT

The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.


Subject(s)
Cell Differentiation , Cell Lineage , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Morphogenesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction , Stem Cells/pathology , Transcription Factors/genetics , Tumor Microenvironment
3.
EMBO J ; 40(11): e107333, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33950524

ABSTRACT

To examine global changes in breast heterogeneity across different states, we determined the single-cell transcriptomes of > 340,000 cells encompassing normal breast, preneoplastic BRCA1+/- tissue, the major breast cancer subtypes, and pairs of tumors and involved lymph nodes. Elucidation of the normal breast microenvironment revealed striking changes in the stroma of post-menopausal women. Single-cell profiling of 34 treatment-naive primary tumors, including estrogen receptor (ER)+ , HER2+ , and triple-negative breast cancers, revealed comparable diversity among cancer cells and a discrete subset of cycling cells. The transcriptomes of preneoplastic BRCA1+/- tissue versus tumors highlighted global changes in the immune microenvironment. Within the tumor immune landscape, proliferative CD8+ T cells characterized triple-negative and HER2+ cancers but not ER+ tumors, while all subtypes comprised cycling tumor-associated macrophages, thus invoking potentially different immunotherapy targets. Copy number analysis of paired ER+ tumors and lymph nodes indicated seeding by genetically distinct clones or mass migration of primary tumor cells into axillary lymph nodes. This large-scale integration of patient samples provides a high-resolution map of cell diversity in normal and cancerous human breast.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Mammary Glands, Human/metabolism , Single-Cell Analysis , Breast Neoplasms/classification , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Female , Gene Expression Profiling , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/pathology , RNA-Seq , Tumor Microenvironment
4.
Breast Cancer Res ; 24(1): 100, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36581993

ABSTRACT

BACKGROUND: After many years of neglect in the field of alternative splicing, the importance of intron retention (IR) in cancer has come into focus following landmark discoveries of aberrant IR patterns in cancer. Many solid and liquid tumours are associated with drastic increases in IR, and such patterns have been pursued as both biomarkers and therapeutic targets. Paradoxically, breast cancer (BrCa) is the only tumour type in which IR is reduced compared to adjacent normal breast tissue. METHODS: In this study, we have conducted a pan-cancer analysis of IR with emphasis on BrCa and its subtypes. We explored mechanisms that could cause aberrant and pathological IR and clarified why normal breast tissue has unusually high IR. RESULTS: Strikingly, we found that aberrantly decreasing IR in BrCa can be largely attributed to normal breast tissue having the highest occurrence of IR events compared to other healthy tissues. Our analyses suggest that low numbers of IR events in breast tumours are associated with poor prognosis, particularly in the luminal B subtype. Interestingly, we found that IR frequencies negatively correlate with cell proliferation in BrCa cells, i.e. rapidly dividing tumour cells have the lowest number of IR events. Aberrant RNA-binding protein expression and changes in tissue composition are among the causes of aberrantly decreasing IR in BrCa. CONCLUSIONS: Our results suggest that IR should be considered for therapeutic manipulation in BrCa patients with aberrantly low IR levels and that further work is needed to understand the cause and impact of high IR in other tumour types.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Introns/genetics , Breast/pathology , Cell Proliferation
5.
Proc Natl Acad Sci U S A ; 116(39): 19530-19540, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31488716

ABSTRACT

Emerging evidence suggests that hepatocytes are primarily maintained by self-renewal during normal liver homeostasis, as well as in response to a wide variety of hepatic injuries. However, how hepatocytes in distinct anatomic locations within the liver lobule are replenished under homeostasis and injury-induced regeneration remains elusive. Using a newly developed bacterial artificial chromosome (BAC)-transgenic mouse model, we demonstrate that Lgr5 expression in the liver is restricted to a unique subset of hepatocytes most adjacent to the central veins. Genetic lineage tracing revealed that pericentral Lgr5+ hepatocytes have a long lifespan and mainly contribute to their own lineage maintenance during postnatal liver development and homeostasis. Remarkably, these hepatocytes also fuel the regeneration of their own lineage during the massive and rapid regeneration process following two-thirds partial hepatectomy. Moreover, Lgr5+ hepatocytes are found to be the main cellular origin of diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) and are highly susceptible to neoplastic transformation triggered by activation of Erbb pathway. Our findings establish an unexpected self-maintaining mode for a defined subset of hepatocytes during liver homeostasis and regeneration, and identify Lgr5+ pericentral hepatocytes as major cells of origin in HCC development.


Subject(s)
Hepatocytes/physiology , Liver Regeneration/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Disease Models, Animal , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/physiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Stem Cells/cytology
6.
Genes Dev ; 28(11): 1143-58, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24888586

ABSTRACT

The mammary epithelium is highly responsive to local and systemic signals, which orchestrate morphogenesis of the ductal tree during puberty and pregnancy. Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. Lineage tracing has highlighted the existence of bipotent mammary stem cells (MaSCs) in situ as well as long-lived unipotent cells that drive morphogenesis and homeostasis of the ductal tree. Moreover, there is accumulating evidence for a heterogeneous MaSC compartment comprising fetal MaSCs, slow-cycling cells, and both long-term and short-term repopulating cells. In parallel, diverse luminal progenitor subtypes have been identified in mouse and human mammary tissue. Elucidation of the normal cellular hierarchy is an important step toward understanding the "cells of origin" and molecular perturbations that drive breast cancer.


Subject(s)
Cell Differentiation , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Stem Cells/cytology , Animals , Cell Lineage , Female , Humans , Stem Cell Transplantation
7.
J Mammary Gland Biol Neoplasia ; 26(1): 71-85, 2021 03.
Article in English | MEDLINE | ID: mdl-33835387

ABSTRACT

Despite rapid advances in our knowledge of the cellular heterogeneity and molecular regulation of the mammary gland, how these relate to 3D cellular organization remains unclear. In addition to hormonal regulation, mammary gland development and function is directed by para- and juxtacrine signaling among diverse cell-types, particularly the immune and mesenchymal populations. Precise mapping of the cellular landscape of the breast will help to decipher this complex coordination. Imaging of thin tissue sections has provided foundational information about cell positioning in the mammary gland and now technological advances in tissue clearing and subcellular-resolution 3D imaging are painting a more complete picture. In particular, confocal, light-sheet and multiphoton microscopy applied to intact tissue can fully capture cell morphology, position and interactions, and have the power to identify spatially rare events. This review will summarize our current understanding of mammary gland cellular organization as revealed by microscopy. We focus on the mouse mammary gland and cover a broad range of immune and stromal cell types at major developmental stages and give insights into important tissue niches and cellular interactions.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Microscopy/methods , Animals , Embryonic Development/physiology , Female , Imaging, Three-Dimensional/methods , Intravital Microscopy/methods , Lactation/physiology , Mice , Microscopy, Confocal/methods , Pregnancy/physiology , Puberty/physiology , Single-Cell Analysis/methods
8.
Breast Cancer Res ; 23(1): 69, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34187545

ABSTRACT

BACKGROUND: Heterogeneity within the mouse mammary epithelium and potential lineage relationships have been recently explored by single-cell RNA profiling. To further understand how cellular diversity changes during mammary ontogeny, we profiled single cells from nine different developmental stages spanning late embryogenesis, early postnatal, prepuberty, adult, mid-pregnancy, late-pregnancy, and post-involution, as well as the transcriptomes of micro-dissected terminal end buds (TEBs) and subtending ducts during puberty. METHODS: The single cell transcriptomes of 132,599 mammary epithelial cells from 9 different developmental stages were determined on the 10x Genomics Chromium platform, and integrative analyses were performed to compare specific time points. RESULTS: The mammary rudiment at E18.5 closely aligned with the basal lineage, while prepubertal epithelial cells exhibited lineage segregation but to a less differentiated state than their adult counterparts. Comparison of micro-dissected TEBs versus ducts showed that luminal cells within TEBs harbored intermediate expression profiles. Ductal basal cells exhibited increased chromatin accessibility of luminal genes compared to their TEB counterparts suggesting that lineage-specific chromatin is established within the subtending ducts during puberty. An integrative analysis of five stages spanning the pregnancy cycle revealed distinct stage-specific profiles and the presence of cycling basal, mixed-lineage, and 'late' alveolar intermediates in pregnancy. Moreover, a number of intermediates were uncovered along the basal-luminal progenitor cell axis, suggesting a continuum of alveolar-restricted progenitor states. CONCLUSIONS: This extended single cell transcriptome atlas of mouse mammary epithelial cells provides the most complete coverage for mammary epithelial cells during morphogenesis to date. Together with chromatin accessibility analysis of TEB structures, it represents a valuable framework for understanding developmental decisions within the mouse mammary gland.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Transcriptome , Animals , Cell Lineage , Chromatin Assembly and Disassembly , Epithelial Cells/cytology , Mammary Glands, Animal/cytology , Mice , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism
9.
PLoS Biol ; 16(8): e2004986, 2018 08.
Article in English | MEDLINE | ID: mdl-30080881

ABSTRACT

Distinct transcriptional states are maintained through organization of chromatin, resulting from the sum of numerous repressive and active histone modifications, into tightly packaged heterochromatin versus more accessible euchromatin. Polycomb repressive complex 2 (PRC2) is the main mammalian complex responsible for histone 3 lysine 27 trimethylation (H3K27me3) and is integral to chromatin organization. Using in vitro and in vivo studies, we show that deletion of Suz12, a core component of all PRC2 complexes, results in loss of H3K27me3 and H3K27 dimethylation (H3K27me2), completely blocks normal mammary gland development, and profoundly curtails progenitor activity in 3D organoid cultures. Through the application of mammary organoids to bypass the severe phenotype associated with Suz12 loss in vivo, we have explored gene expression and chromatin structure in wild-type and Suz12-deleted basal-derived organoids. Analysis of organoids led to the identification of lineage-specific changes in gene expression and chromatin structure, inferring cell type-specific PRC2-mediated gene silencing of the chromatin state. These expression changes were accompanied by cell cycle arrest but not lineage infidelity. Together, these data indicate that canonical PRC2 function is essential for development of the mammary gland through the repression of alternate transcription programs and maintenance of chromatin states.


Subject(s)
Mammary Glands, Animal/embryology , Polycomb Repressive Complex 2/physiology , Animals , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/physiology , Female , Heterochromatin/metabolism , Histone Code , Histones/metabolism , Lysine/metabolism , Mammary Glands, Animal/metabolism , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins , Primary Cell Culture , Protein Processing, Post-Translational
10.
Development ; 144(6): 1065-1071, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27993977

ABSTRACT

Advances in stem cell research have enabled the generation of 'mini organs' or organoids that recapitulate phenotypic traits of the original biological specimen. Although organoids have been demonstrated for multiple organ systems, there are more limited options for studying mouse mammary gland formation in vitro Here, we have built upon previously described culture assays to define culture conditions that enable the efficient generation of clonal organoid structures from single sorted basal mammary epithelial cells (MECs). Analysis of Confetti-reporter mice revealed the formation of uni-colored structures and thus the clonal nature of these organoids. High-resolution 3D imaging demonstrated that basal cell-derived complex organoids comprised an inner compartment of polarized luminal cells with milk-producing capacity and an outer network of elongated myoepithelial cells. Conversely, structures generated from luminal MECs rarely contained basal/myoepithelial cells. Moreover, flow cytometry and 3D microscopy of organoids generated from lineage-specific reporter mice established the bipotent capacity of basal cells and the restricted potential of luminal cells. In summary, we describe optimized in vitro conditions for the efficient generation of mouse mammary organoids that recapitulate features of mammary tissue architecture and function, and can be applied to understand tissue dynamics and cell-fate decisions.


Subject(s)
Mammary Glands, Animal/growth & development , Organoids/cytology , Tissue Culture Techniques/methods , Animals , Cell Lineage , Clone Cells , Epithelial Cells/cytology , Female , Flow Cytometry , Genes, Reporter , Imaging, Three-Dimensional , Mammary Glands, Animal/cytology , Mice , Microscopy, Confocal
11.
Nature ; 506(7488): 322-7, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24463516

ABSTRACT

The mammary epithelium undergoes profound morphogenetic changes during development. Architecturally, it comprises two primary lineages, the inner luminal and outer myoepithelial cell layers. Two opposing concepts on the nature of mammary stem cells (MaSCs) in the postnatal gland have emerged. One model, based on classical transplantation assays, postulates that bipotent MaSCs have a key role in coordinating ductal epithelial expansion and maintenance in the adult gland, whereas the second model proposes that only unipotent MaSCs identified by lineage tracing contribute to these processes. Through clonal cell-fate mapping studies using a stochastic multicolour cre reporter combined with a new three-dimensional imaging strategy, we provide evidence for the existence of bipotent MaSCs as well as distinct long-lived progenitor cells. The cellular dynamics at different developmental stages support a model in which both stem and progenitor cells drive morphogenesis during puberty, whereas bipotent MaSCs coordinate ductal homeostasis and remodelling of the mouse adult gland.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Multipotent Stem Cells/cytology , Animals , Cell Lineage , Cell Tracking , Clone Cells/cytology , Clone Cells/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Keratin-14/metabolism , Mice , Morphogenesis , Multipotent Stem Cells/metabolism , Puberty , Receptors, G-Protein-Coupled/metabolism , Sexual Maturation , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Blood ; 130(23): 2504-2515, 2017 12 07.
Article in English | MEDLINE | ID: mdl-28972012

ABSTRACT

T-cell differentiation is governed by interactions with thymic epithelial cells (TECs) and defects in this process undermine immune function and tolerance. To uncover new strategies to restore thymic function and adaptive immunity in immunodeficiency, we sought to determine the molecular mechanisms that control life and death decisions in TECs. Guided by gene expression profiling, we created mouse models that specifically deleted prosurvival genes in TECs. We found that although BCL-2 and BCL-XL were dispensable for TEC homeostasis, MCL-1 deficiency impacted on TECs as early as embryonic day 15.5, resulting in early thymic atrophy and T-cell lymphopenia, with near complete loss of thymic tissue by 2 months of age. MCL-1 was not necessary for TEC differentiation but was continually required for the survival of mature cortical and medullary TECs and the maintenance of thymic architecture. A screen of TEC trophic factors in organ cultures showed that epidermal growth factor upregulated MCL-1 via MAPK/ERK kinase activity, providing a molecular mechanism for the support of TEC survival. This signaling axis governing TEC survival and thymic function represents a new target for strategies for thymic protection and regeneration.


Subject(s)
Cell Survival/genetics , Epithelial Cells/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Thymus Gland/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Homeostasis/genetics , Immunophenotyping , Lymphopenia/genetics , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/pathology , bcl-X Protein/genetics , bcl-X Protein/metabolism
13.
Dev Biol ; 424(1): 77-89, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28215940

ABSTRACT

The mammalian kidney collecting ducts are critical for water, electrolyte and acid-base homeostasis and develop as a branched network of tubular structures composed of principal cells intermingled with intercalated cells. The intermingled nature of the different collecting duct cell types has made it challenging to identify unique and critical factors that mark and/or regulate the development of the different collecting duct cell lineages. Here we report that the canonical Notch signaling pathway components, RBPJ and Presinilin1 and 2, are involved in patterning the mouse collecting duct cell fates by maintaining a balance between principal cell and intercalated cell fates. The relatively reduced number of principal cells in Notch-signaling-deficient kidneys offered a unique genetic leverage to identify critical principal cell-enriched factors by transcriptional profiling. Elf5, which codes for an ETS transcription factor, is one such gene that is down-regulated in kidneys with Notch-signaling-deficient collecting ducts. Additionally, Elf5 is among the earliest genes up regulated by ectopic expression of activated Notch1 in the developing collecting ducts. In the kidney, Elf5 is first expressed early within developing collecting ducts and remains on in mature principal cells. Lineage tracing of Elf5-expressing cells revealed that they are committed to the principal cell lineage by as early as E16.5. Over-expression of ETS Class IIa transcription factors, including Elf5, Elf3 and Ehf, increase the transcriptional activity of the proximal promoters of Aqp2 and Avpr2 in cultured ureteric duct cell lines. Conditional inactivation of Elf5 in the developing collecting ducts results in a small but significant reduction in the expression levels of Aqp2 and Avpr2 genes. We have identified Elf5 as an early maker of the principal cell lineage that contributes to the expression of principal cell specific genes.


Subject(s)
Aquaporin 2/genetics , Cell Lineage , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Kidney/cytology , Kidney/metabolism , Receptors, Vasopressin/genetics , Transcription Factors/metabolism , Animals , Aquaporin 2/metabolism , Cell Count , Cell Line , Down-Regulation/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Integrases/metabolism , Kidney/embryology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/metabolism , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptors, Notch/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction , Up-Regulation/genetics , Ureter/embryology , Ureter/metabolism
14.
Stem Cells ; 35(3): 626-640, 2017 03.
Article in English | MEDLINE | ID: mdl-28009074

ABSTRACT

The study and application of human pluripotent stem cells (hPSCs) will be enhanced by the availability of well-characterized monoclonal antibodies (mAbs) detecting cell-surface epitopes. Here, we report generation of seven new mAbs that detect cell surface proteins present on live and fixed human ES cells (hESCs) and human iPS cells (hiPSCs), confirming our previous prediction that these proteins were present on the cell surface of hPSCs. The mAbs all show a high correlation with POU5F1 (OCT4) expression and other hPSC surface markers (TRA-160 and SSEA-4) in hPSC cultures and detect rare OCT4 positive cells in differentiated cell cultures. These mAbs are immunoreactive to cell surface protein epitopes on both primed and naive state hPSCs, providing useful research tools to investigate the cellular mechanisms underlying human pluripotency and states of cellular reprogramming. In addition, we report that subsets of the seven new mAbs are also immunoreactive to human bone marrow-derived mesenchymal stem cells (MSCs), normal human breast subsets and both normal and tumorigenic colorectal cell populations. The mAbs reported here should accelerate the investigation of the nature of pluripotency, and enable development of robust cell separation and tracing technologies to enrich or deplete for hPSCs and other human stem and somatic cell types. Stem Cells 2017;35:626-640.


Subject(s)
Antibodies, Monoclonal/immunology , Membrane Proteins/immunology , Pluripotent Stem Cells/metabolism , Animals , Antigens, Surface/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Self Renewal , Down-Regulation/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/metabolism , Mice , Octamer Transcription Factor-3/metabolism
15.
Cancer Metastasis Rev ; 35(4): 547-573, 2016 12.
Article in English | MEDLINE | ID: mdl-28025748

ABSTRACT

Patient-derived xenograft (PDX) models of a growing spectrum of cancers are rapidly supplanting long-established traditional cell lines as preferred models for conducting basic and translational preclinical research. In breast cancer, to complement the now curated collection of approximately 45 long-established human breast cancer cell lines, a newly formed consortium of academic laboratories, currently from Europe, Australia, and North America, herein summarizes data on over 500 stably transplantable PDX models representing all three clinical subtypes of breast cancer (ER+, HER2+, and "Triple-negative" (TNBC)). Many of these models are well-characterized with respect to genomic, transcriptomic, and proteomic features, metastatic behavior, and treatment response to a variety of standard-of-care and experimental therapeutics. These stably transplantable PDX lines are generally available for dissemination to laboratories conducting translational research, and contact information for each collection is provided. This review summarizes current experiences related to PDX generation across participating groups, efforts to develop data standards for annotation and dissemination of patient clinical information that does not compromise patient privacy, efforts to develop complementary data standards for annotation of PDX characteristics and biology, and progress toward "credentialing" of PDX models as surrogates to represent individual patients for use in preclinical and co-clinical translational research. In addition, this review highlights important unresolved questions, as well as current limitations, that have hampered more efficient generation of PDX lines and more rapid adoption of PDX use in translational breast cancer research.


Subject(s)
Breast Neoplasms/pathology , Disease Models, Animal , Animals , Female , Heterografts , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Translational Research, Biomedical
16.
Development ; 141(16): 3159-64, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25038044

ABSTRACT

The HLH transcriptional regulator Id4 exerts important roles in different organs, including the neural compartment, where Id4 loss usually results in early lethality. To explore the role of this basally restricted transcription factor in the mammary gland, we generated a cre-inducible mouse model. MMTV- or K14-cre-mediated deletion of Id4 led to a delay in ductal morphogenesis, consistent with previous findings using a germ-line knockout mouse model. A striking increase in the expression of ERα (Esr1), PR and FoxA1 was observed in both the basal and luminal cellular subsets of Id4-deficient mammary glands. Together with chromatin immunoprecipitation of Id4 on the Esr1 and Foxa1 promoter regions, these data imply that Id4 is a negative regulator of the ERα signaling axis. Unexpectedly, examination of the ovaries of targeted mice revealed significantly increased numbers of secondary and antral follicles, and reduced Id4 expression in the granulosa cells. Moreover, expression of the cascade of enzymes that are crucial for estrogen biosynthesis in the ovary was decreased in Id4-deficient females and uterine weights were considerably lower, indicating impaired estrogen production. Thus, compromised ovarian function and decreased circulating estrogen likely contribute to the mammary ductal defects evident in Id4-deficient mice. Collectively, these data identify Id4 as a novel regulator of estrogen signaling, where Id4 restrains ERα expression in the basal and luminal cellular compartments of the mammary gland and regulates estrogen biosynthesis in the ovary.


Subject(s)
Estrogens/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/physiology , Mammary Glands, Animal/physiology , Ovary/physiology , Animals , Base Sequence , Estrogen Receptor alpha/metabolism , Female , Gene Deletion , Gene Expression Regulation , Granulosa Cells/cytology , Hepatocyte Nuclear Factor 3-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Progesterone/metabolism , Signal Transduction , Uterus/physiology
17.
Nature ; 469(7330): 314-22, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21248838

ABSTRACT

Both solid tumours and leukaemias show considerable histological and functional heterogeneity. It is widely accepted that genetic lesions have a major role in determining tumour phenotype, but evidence is also accumulating that cancers of distinct subtypes within an organ may derive from different 'cells of origin'. These cells acquire the first genetic hit or hits that culminate in the initiation of cancer. The identification of these crucial target cell populations may allow earlier detection of malignancies and better prediction of tumour behaviour, and ultimately may lead to preventive therapies for individuals at high risk of developing cancer.


Subject(s)
Cell Lineage , Neoplasms/pathology , Animals , Hematologic Neoplasms/pathology , Humans , Models, Biological , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Neoplastic Stem Cells/pathology , Phenotype
18.
PLoS Genet ; 10(5): e1004323, 2014 May.
Article in English | MEDLINE | ID: mdl-24852022

ABSTRACT

Polarity coordinates cell movement, differentiation, proliferation and apoptosis to build and maintain complex epithelial tissues such as the mammary gland. Loss of polarity and the deregulation of these processes are critical events in malignant progression but precisely how and at which stage polarity loss impacts on mammary development and tumourigenesis is unclear. Scrib is a core polarity regulator and tumour suppressor gene however to date our understanding of Scrib function in the mammary gland has been limited to cell culture and transplantation studies of cell lines. Utilizing a conditional mouse model of Scrib loss we report for the first time that Scrib is essential for mammary duct morphogenesis, mammary progenitor cell fate and maintenance, and we demonstrate a critical and specific role for Scribble in the control of the early steps of breast cancer progression. In particular, Scrib-deficiency significantly induced Fra1 expression and basal progenitor clonogenicity, which resulted in fully penetrant ductal hyperplasia characterized by high cell turnover, MAPK hyperactivity, frank polarity loss with mixing of apical and basolateral membrane constituents and expansion of atypical luminal cells. We also show for the first time a role for Scribble in mammalian spindle orientation with the onset of mammary hyperplasia being associated with aberrant luminal cell spindle orientation and a failure to apoptose during the final stage of duct tubulogenesis. Restoring MAPK/Fra1 to baseline levels prevented Scrib-hyperplasia, whereas persistent Scrib deficiency induced alveolar hyperplasia and increased the incidence, onset and grade of mammary tumours. These findings, based on a definitive genetic mouse model provide fundamental insights into mammary duct maturation and homeostasis and reveal that Scrib loss activates a MAPK/Fra1 pathway that alters mammary progenitor activity to drive premalignancy and accelerate tumour progression.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , MAP Kinase Signaling System , Mammary Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cell Polarity , Female , Homeostasis , Hyperplasia , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Morphogenesis
19.
Genes Dev ; 23(22): 2563-77, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19933147

ABSTRACT

The epithelium of the mammary gland exists in a highly dynamic state, undergoing dramatic morphogenetic changes during puberty, pregnancy, lactation, and regression. The recent identification of stem and progenitor populations in mouse and human mammary tissue has provided evidence that the mammary epithelium is organized in a hierarchical manner. Characterization of these normal epithelial subtypes is an important step toward understanding which cells are predisposed to oncogenesis. This review summarizes progress in the field toward defining constituent cells and key molecular regulators of the mammary epithelial hierarchy. Potential relationships between normal epithelial populations and breast tumor subtypes are discussed, with implications for understanding the cellular etiology underpinning breast tumor heterogeneity.


Subject(s)
Breast Neoplasms/pathology , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Differentiation , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/transplantation , Mammary Glands, Human/cytology , Mutation/genetics , Pregnancy , Stem Cells/cytology , Ubiquitin-Protein Ligases/genetics , Wnt Proteins/metabolism
20.
Biochim Biophys Acta ; 1853(7): 1683-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25882817

ABSTRACT

The Ste20-like kinase SLK plays a pivotal role in cell migration and focal adhesion turnover and is regulated by the LIM domain-binding proteins Ldb1 and Ldb2. These adapter proteins have been demonstrated to interact with LMO4 in the organization of transcriptional complexes. Therefore, we have assessed the ability of LMO4 to also interact and regulate SLK activity. Our data show that LMO4 can directly bind to SLK and activate its kinase activity in vitro and in vivo. LMO4 can be co-precipitated with SLK following the induction of cell migration by scratch wounding and Cre-mediated deletion of LMO4 in conditional LMO4(fl/fl) fibroblasts inhibits cell migration and SLK activation. Deletion of LMO4 impairs Ldb1 and SLK recruitment to the leading edge of migrating cells. Supporting this, Src/Yes/Fyn-deficient cells (SYF) expressing very low levels of LMO4 do not recruit SLK to the leading edge. Re-expression of wildtype Myc-LMO4 in SYF cells, but not a mutant version, restores SLK localization and kinase activity. Overall, our data suggest that activation of SLK by haptotactic signals requires its recruitment to the leading edge by LMO4 in a Src-dependent manner. Furthermore, this establishes a novel cytosolic role for the transcriptional co-activator LMO4.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Fibroblasts/cytology , Fibroblasts/enzymology , LIM Domain Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Pseudopodia/metabolism , src-Family Kinases/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing/chemistry , Animals , DNA-Binding Proteins/metabolism , Enzyme Activation , Female , Gene Deletion , HEK293 Cells , Humans , LIM Domain Proteins/chemistry , Mice , Protein Binding , Protein Structure, Tertiary , Protein Transport , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL