61 results on '"Garbe JC"'
Search Results
2. An Integrated Human Mammary epithelial Cell Culture System for Studying Carcinogenesis and Aging
- Author
-
Stampfer, MR, LaBarge, MA, and Garbe, JC
- Published
- 2018
3. 184AA3: a xenograft model of ER+ breast adenocarcinoma
- Author
-
Hines, WC, Kuhn, I, Thi, K, Chu, B, Stanford-Moore, G, Sampayo, R, Garbe, JC, Stampfer, M, Borowsky, AD, and Bissell, MJ
- Subjects
Clinical Sciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
Despite the prevalence and significant morbidity resulting from estrogen receptor positive (ER+) breast adenocarcinomas, there are only a few models of this cancer subtype available for drug development and arguably none for studying etiology. Those models that do exist have questionable clinical relevance. Given our goal of developing luminal models, we focused on six cell lines derived by minimal mutagenesis from normal human breast cells, and asked if any could generate clinically relevant xenografts, which we then extensively characterized. Xenografts of one cell line, 184AA3, consistently formed ER+ adenocarcinomas that had a high proliferative rate and other features consistent with “luminal B” intrinsic subtype. Squamous and spindle cell/mesenchymal differentiation was absent, in stark contrast to other cell lines that we examined or others have reported. We explored intratumoral heterogeneity produced by 184AA3 by immunophenotyping xenograft tumors and cultured cells, and characterized marker expression by immunofluorescence and flow cytometry. A CD44High subpopulation was discovered, yet their tumor forming ability was far less than CD44Low cells. Single cell cloning revealed the phenotypic plasticity of 184AA3, consistent with the intratumoral heterogeneity observed in xenografts. Characterization of ER expression in cultures revealed ER protein and signaling is intact, yet when estrogen was depleted in culture, and in vivo, it did not impact cell or tumor growth, analogous to therapeutically resistant ER+ cancers. This model is appropriate for studies of the etiology of ovarian hormone independent adenocarcinomas, for identification of therapeutic targets, predictive testing, and drug development.
- Published
- 2016
4. Programmed synthesis of three-dimensional tissues
- Author
-
Gartner, Zev, Desai, Tejal, Todhunter, ME, Jee, NY, Hughes, AJ, Coyle, MC, Cerchiari, A, Farlow, J, Garbe, JC, LaBarge, MA, Desai, TA, and Gartner, ZJ
- Abstract
© 2015 Nature America, Inc. All rights reserved.Reconstituting tissues from their cellular building blocks facilitates the modeling of morphogenesis, homeostasis and disease in vitro. Here we describe DNA-programmed assembly of cells (DPAC), a method to re
- Published
- 2015
5. An Integrated Human Mammary epithelial Cell Culture System for Studying Carcinogenesis and Aging
- Author
-
Stampfer, MR, Stampfer, MR, LaBarge, MA, Garbe, JC, Stampfer, MR, Stampfer, MR, LaBarge, MA, and Garbe, JC
- Published
- 2022
6. Rapid Organoid Reconstitution by Chemical Micromolding
- Author
-
Weber, RJ, Cerchiari, AE, Delannoy, LS, Garbe, JC, Labarge, MA, Desai, TA, and Gartner, ZJ
- Subjects
DNA programmed assembly ,mammary gland ,1.1 Normal biological development and functioning ,organoid ,Biomedical Engineering ,Bioengineering ,Generic health relevance ,synthetic biology ,3D tissue culture ,microwell ,Biotechnology - Abstract
© 2016 American Chemical Society. Purified populations of cells can be reconstituted into organoids that recapitulate aspects of their in vivo structure and function. These organoids are useful as models of healthy and diseased tissue in the basic sciences, in vitro screens, and regenerative medicine. Existing strategies to reconstitute organoids from purified cells face obstacles with respect to cell-viability, multicellular connectivity, scalability, and compatibility with subsequent experimental or analytical techniques. To address these challenges, we developed a strategy for rapidly casting populations of cells into microtissues of prescribed size and shape. This approach begins by chemically remodeling the adhesive properties of living cells with membrane-anchored ssDNA with modest annealing kinetics. Populations of complementary labeled cells are then combined into microwells that rapidly mold the DNA-adhesive cell populations into 3D aggregates of uniform size and shape. Once formed, aggregates are removed from the molds in the presence of "capping" oligonucleotides that block hybridization of residual surface DNA between aggregates in suspension. Finally, transfer of aggregates to biomimetic gels for 3D culture completes the process of reconstitution. This strategy of chemical micromolding allows for control over aggregate internal topology and does not perturb the natural process of self-organization in primary human mammary epithelial cells.
- Published
- 2016
- Full Text
- View/download PDF
7. An expedited screening platform for the discovery of anti-ageing compounds in vitro and in vivo.
- Author
-
Lujan C, Tyler EJ, Ecker S, Webster AP, Stead ER, Martinez-Miguel VE, Milligan D, Garbe JC, Stampfer MR, Beck S, Lowe R, Bishop CL, and Bjedov I
- Subjects
- Humans, Animals, Epigenesis, Genetic drug effects, Drug Discovery methods, Cellular Senescence drug effects, Drug Evaluation, Preclinical methods, Drosophila, Cells, Cultured, Sirolimus pharmacology, DNA Methylation drug effects, Longevity drug effects, Aging drug effects
- Abstract
Background: Restraining or slowing ageing hallmarks at the cellular level have been proposed as a route to increased organismal lifespan and healthspan. Consequently, there is great interest in anti-ageing drug discovery. However, this currently requires laborious and lengthy longevity analysis. Here, we present a novel screening readout for the expedited discovery of compounds that restrain ageing of cell populations in vitro and enable extension of in vivo lifespan., Methods: Using Illumina methylation arrays, we monitored DNA methylation changes accompanying long-term passaging of adult primary human cells in culture. This enabled us to develop, test, and validate the CellPopAge Clock, an epigenetic clock with underlying algorithm, unique among existing epigenetic clocks for its design to detect anti-ageing compounds in vitro. Additionally, we measured markers of senescence and performed longevity experiments in vivo in Drosophila, to further validate our approach to discover novel anti-ageing compounds. Finally, we bench mark our epigenetic clock with other available epigenetic clocks to consolidate its usefulness and specialisation for primary cells in culture., Results: We developed a novel epigenetic clock, the CellPopAge Clock, to accurately monitor the age of a population of adult human primary cells. We find that the CellPopAge Clock can detect decelerated passage-based ageing of human primary cells treated with rapamycin or trametinib, well-established longevity drugs. We then utilise the CellPopAge Clock as a screening tool for the identification of compounds which decelerate ageing of cell populations, uncovering novel anti-ageing drugs, torin2 and dactolisib (BEZ-235). We demonstrate that delayed epigenetic ageing in human primary cells treated with anti-ageing compounds is accompanied by a reduction in senescence and ageing biomarkers. Finally, we extend our screening platform in vivo by taking advantage of a specially formulated holidic medium for increased drug bioavailability in Drosophila. We show that the novel anti-ageing drugs, torin2 and dactolisib (BEZ-235), increase longevity in vivo., Conclusions: Our method expands the scope of CpG methylation profiling to accurately and rapidly detecting anti-ageing potential of drugs using human cells in vitro, and in vivo, providing a novel accelerated discovery platform to test sought after anti-ageing compounds and geroprotectors., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. Author Correction: The senescent methylome and its relationship with cancer, ageing and germline genetic variation in humans.
- Author
-
Lowe R, Overhoff MG, Ramagopalan SV, Garbe JC, Koh J, Stampfer MR, Beach DH, Rakyan VK, and Bishop CL
- Published
- 2024
- Full Text
- View/download PDF
9. Configurational entropy is an intrinsic driver of tissue structural heterogeneity.
- Author
-
Srivastava V, Hu JL, Garbe JC, Veytsman B, Shalabi SF, Yllanes D, Thomson M, LaBarge MA, Huber G, and Gartner ZJ
- Abstract
Tissues comprise ordered arrangements of cells that can be surprisingly disordered in their details. How the properties of single cells and their microenvironment contribute to the balance between order and disorder at the tissue-scale remains poorly understood. Here, we address this question using the self-organization of human mammary organoids as a model. We find that organoids behave like a dynamic structural ensemble at the steady state. We apply a maximum entropy formalism to derive the ensemble distribution from three measurable parameters - the degeneracy of structural states, interfacial energy, and tissue activity (the energy associated with positional fluctuations). We link these parameters with the molecular and microenvironmental factors that control them to precisely engineer the ensemble across multiple conditions. Our analysis reveals that the entropy associated with structural degeneracy sets a theoretical limit to tissue order and provides new insight for tissue engineering, development, and our understanding of disease progression., Competing Interests: DECLARATION OF INTERESTS Z.J.G. is an equity holder in Scribe biosciences, Provenance Bio, and Serotiny.
- Published
- 2023
- Full Text
- View/download PDF
10. Evidence for accelerated aging in mammary epithelia of women carrying germline BRCA1 or BRCA2 mutations.
- Author
-
Shalabi SF, Miyano M, Sayaman RW, Lopez JC, Jokela TA, Todhunter ME, Hinz S, Garbe JC, Stampfer MR, Kessenbrock K, Seewaldt VE, and LaBarge MA
- Subjects
- Humans, Female, Aging genetics, Breast pathology, Germ-Line Mutation genetics, BRCA1 Protein genetics, BRCA2 Protein genetics, Mammary Glands, Human, Breast Neoplasms genetics
- Abstract
During aging in the human mammary gland, luminal epithelial cells lose lineage fidelity by expressing markers normally expressed in myoepithelial cells. We hypothesize that loss of lineage fidelity is a general manifestation of epithelia that are susceptible to cancer initiation. In the present study, we show that histologically normal breast tissue from younger women who are susceptible to breast cancer, as a result of harboring a germline mutation in BRCA1 , BRCA2 or PALB2 genes, exhibits hallmarks of accelerated aging. These include proportionately increased luminal epithelial cells that acquired myoepithelial markers, decreased proportions of myoepithelial cells and a basal differentiation bias or failure of differentiation of cKit
+ progenitors. High-risk luminal and myoepithelial cells are transcriptionally enriched for genes of the opposite lineage, inflammatory- and cancer-related pathways. We have identified breast-aging hallmarks that reflect a convergent biology of cancer susceptibility, regardless of the specific underlying genetic or age-dependent risk or the associated breast cancer subtype., Competing Interests: Competing interests The authors declare no competing interests.- Published
- 2021
- Full Text
- View/download PDF
11. Early growth response 2 (EGR2) is a novel regulator of the senescence programme.
- Author
-
Tyler EJ, Gutierrez Del Arroyo A, Hughes BK, Wallis R, Garbe JC, Stampfer MR, Koh J, Lowe R, Philpott MP, and Bishop CL
- Subjects
- Adolescent, Adult, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Epithelial Cells cytology, Epithelial Cells metabolism, Fibroblasts cytology, Fibroblasts metabolism, Gene Knockdown Techniques, Humans, Mammary Glands, Human cytology, Protein Binding, RNA, Small Interfering metabolism, Retinoblastoma Protein metabolism, Tumor Suppressor Protein p53 metabolism, Up-Regulation, Young Adult, Cellular Senescence, Early Growth Response Protein 2 metabolism
- Abstract
Senescence, a state of stable growth arrest, plays an important role in ageing and age-related diseases in vivo. Although the INK4/ARF locus is known to be essential for senescence programmes, the key regulators driving p16 and ARF transcription remain largely underexplored. Using siRNA screening for modulators of the p16/pRB and ARF/p53/p21 pathways in deeply senescent human mammary epithelial cells (DS HMECs) and fibroblasts (DS HMFs), we identified EGR2 as a novel regulator of senescence. EGR2 expression is up-regulated during senescence, and its ablation by siRNA in DS HMECs and HMFs transiently reverses the senescent phenotype. We demonstrate that EGR2 activates the ARF and p16 promoters and directly binds to both the ARF and p16 promoters. Loss of EGR2 down-regulates p16 levels and increases the pool of p16- p21- 'reversed' cells in the population. Moreover, EGR2 overexpression is sufficient to induce senescence. Our data suggest that EGR2 is a direct transcriptional activator of the p16/pRB and ARF/p53/p21 pathways in senescence and a novel marker of senescence., (© 2021 The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
12. Different culture media modulate growth, heterogeneity, and senescence in human mammary epithelial cell cultures.
- Author
-
Lee JK, Bloom J, Zubeldia-Plazaola A, Garbe JC, Stampfer MR, and LaBarge MA
- Subjects
- Adult, Cell Lineage physiology, Cells, Cultured, Female, Humans, Mammaplasty methods, Middle Aged, Phenotype, Young Adult, Cellular Senescence physiology, Culture Media metabolism, Epithelial Cells metabolism, Epithelial Cells physiology, Mammary Glands, Human metabolism, Mammary Glands, Human physiology
- Abstract
The ability to culture normal human mammary epithelial cells (HMEC) greatly facilitates experiments that seek to understand both normal mammary cell biology and the many differences between normal and abnormal human mammary epithelia. To maximize in vivo relevance, the primary cell culture conditions should maintain cells in states that resemble in vivo as much as possible. Towards this goal, we compared the properties of HMEC strains from two different reduction mammoplasty tissues that were grown in parallel using different media and culture conditions. Epithelial organoids were initiated into three different media: two commonly used serum-free-media, MCDB 170-type (e.g. MEGM) and WIT-P, and a low stress media, M87A. Growth, lineage heterogeneity, p16 protein expression, and population doublings to senescence were measured for each culture condition. MCDB 170 caused rapid senescence and loss of heterogeneity within 2 to 3 passages, but some cultures went through the 1 to 2 month process of selection to generate clonal finite post-selection post-stasis cells. WIT-P caused impressive expansion of luminal cells in 2nd passage followed by their near complete disappearance by passage 4 and senescence shortly thereafter. M87A supported as much as twice the number of population doublings compared to either serum-free medium, and luminal and myoepithelial cells were present for as many as 8 passages. Thus, of the three media compared, WIT-P and MCDB 170 imposed rapid senescence and loss of lineage heterogeneity, phenotypes consistent with cells maintained in high-stress conditions, while M87A supported cultures that maintained multiple lineages and robust growth for up to 60 population doublings. In conjunction with previous studies examining the molecular properties of cultures grown in these media, we conclude that M87A medium is most able to support long-term culture of multiple lineages similar to in vivo conditions, thereby facilitating investigations of normal HMEC biology relevant to the mammary gland in situ., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
13. Microenvironment-Induced Non-sporadic Expression of the AXL and cKIT Receptors Are Related to Epithelial Plasticity and Drug Resistance.
- Author
-
Jokela TA, Engelsen AST, Rybicka A, Pelissier Vatter FA, Garbe JC, Miyano M, Tiron C, Ferariu D, Akslen LA, Stampfer MR, Lorens JB, and LaBarge MA
- Abstract
The existence of rare cancer cells that sporadically acquire drug-tolerance through epigenetic mechanisms is proposed as one mechanism that drives cancer therapy failure. Here we provide evidence that specific microenvironments impose non-sporadic expression of proteins related to epithelial plasticity and drug resistance. Microarrays of robotically printed combinatorial microenvironments of known composition were used to make cell-based functional associations between microenvironments, which were design-inspired by normal and tumor-burdened breast tissues, and cell phenotypes. We hypothesized that specific combinations of microenvironment constituents non-sporadically impose the induction of the AXL and cKIT receptor tyrosine kinase proteins, which are known to be involved in epithelial plasticity and drug-tolerance, in an isogenic human mammary epithelial cell (HMEC) malignant progression series. Dimension reduction analysis reveals type I collagen as a dominant feature, inducing expression of both markers in pre-stasis finite lifespan HMECs, and transformed non-malignant and malignant immortal cell lines. Basement membrane-associated matrix proteins, laminin-111 and type IV collagen, suppress AXL and cKIT expression in pre-stasis and non-malignant cells. However, AXL and cKIT are not suppressed by laminin-111 in malignant cells. General linear models identified key factors, osteopontin, IL-8, and type VIα3 collagen, which significantly upregulated AXL and cKIT, as well as a plasticity-related gene expression program that is often observed in stem cells and in epithelial-to-mesenchymal-transition. These factors are co-located with AXL-expressing cells in situ in normal and breast cancer tissues, and associated with resistance to paclitaxel. A greater diversity of microenvironments induced AXL and cKIT expression consistent with plasticity and drug-tolerant phenotypes in tumorigenic cells compared to normal or immortal cells, suggesting a reduced perception of microenvironment specificity in malignant cells. Microenvironment-imposed reprogramming could explain why resistant cells are seemingly persistent and rapidly adaptable to multiple classes of drugs. These results support the notion that specific microenvironments drive drug-tolerant cellular phenotypes and suggest a novel interventional avenue for preventing acquired therapy resistance.
- Published
- 2018
- Full Text
- View/download PDF
14. Rapid Organoid Reconstitution by Chemical Micromolding.
- Author
-
Weber RJ, Cerchiari AE, Delannoy LS, Garbe JC, LaBarge MA, Desai TA, and Gartner ZJ
- Abstract
Purified populations of cells can be reconstituted into organoids that recapitulate aspects of their in vivo structure and function. These organoids are useful as models of healthy and diseased tissue in the basic sciences, in vitro screens, and regenerative medicine. Existing strategies to reconstitute organoids from purified cells face obstacles with respect to cell-viability, multicellular connectivity, scalability, and compatibility with subsequent experimental or analytical techniques. To address these challenges, we developed a strategy for rapidly casting populations of cells into microtissues of prescribed size and shape. This approach begins by chemically remodeling the adhesive properties of living cells with membrane-anchored ssDNA with modest annealing kinetics. Populations of complementary labeled cells are then combined into microwells that rapidly mold the DNA-adhesive cell populations into 3D aggregates of uniform size and shape. Once formed, aggregates are removed from the molds in the presence of "capping" oligonucleotides that block hybridization of residual surface DNA between aggregates in suspension. Finally, transfer of aggregates to biomimetic gels for 3D culture completes the process of reconstitution. This strategy of chemical micromolding allows for control over aggregate internal topology and does not perturb the natural process of self-organization in primary human mammary epithelial cells.
- Published
- 2016
- Full Text
- View/download PDF
15. Chromatin remodeling regulates catalase expression during cancer cells adaptation to chronic oxidative stress.
- Author
-
Glorieux C, Sandoval JM, Fattaccioli A, Dejeans N, Garbe JC, Dieu M, Verrax J, Renard P, Huang P, and Calderon PB
- Subjects
- Adaptation, Physiological, Base Sequence, Catalase metabolism, Cell Line, Cell Line, Tumor, Chromatin chemistry, Chromatin drug effects, Epithelial Cells cytology, Epithelial Cells drug effects, Epithelial Cells metabolism, Histone Deacetylases genetics, Histone Deacetylases metabolism, Humans, Hydrogen Peroxide metabolism, Hydrogen Peroxide pharmacology, MCF-7 Cells, Oxidative Stress, Promoter Regions, Genetic, Reactive Oxygen Species antagonists & inhibitors, Reactive Oxygen Species metabolism, Retinoic Acid Receptor alpha metabolism, Signal Transduction, Transcription Factors metabolism, Transcription, Genetic, Catalase genetics, Chromatin metabolism, Chromatin Assembly and Disassembly, Gene Expression Regulation, Neoplastic, Retinoic Acid Receptor alpha genetics, Transcription Factors genetics
- Abstract
Regulation of ROS metabolism plays a major role in cellular adaptation to oxidative stress in cancer cells, but the molecular mechanism that regulates catalase, a key antioxidant enzyme responsible for conversion of hydrogen peroxide to water and oxygen, remains to be elucidated. Therefore, we investigated the transcriptional regulatory mechanism controlling catalase expression in three human mammary cell lines: the normal mammary epithelial 250MK primary cells, the breast adenocarcinoma MCF-7 cells and an experimental model of MCF-7 cells resistant against oxidative stress resulting from chronic exposure to H
2 O2 (Resox), in which catalase was overexpressed. Here we identify a novel promoter region responsible for the regulation of catalase expression at -1518/-1226 locus and the key molecules that interact with this promoter and affect catalase transcription. We show that the AP-1 family member JunB and retinoic acid receptor alpha (RARα) mediate catalase transcriptional activation and repression, respectively, by controlling chromatin remodeling through a histone deacetylases-dependent mechanism. This regulatory mechanism plays an important role in redox adaptation to chronic exposure to H2 O2 in breast cancer cells. Our study suggests that cancer adaptation to oxidative stress may be regulated by transcriptional factors through chromatin remodeling, and reveals a potential new mechanism to target cancer cells., (Copyright © 2016 Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
16. Programmed synthesis of three-dimensional tissues.
- Author
-
Todhunter ME, Jee NY, Hughes AJ, Coyle MC, Cerchiari A, Farlow J, Garbe JC, LaBarge MA, Desai TA, and Gartner ZJ
- Subjects
- Cell Adhesion, Cell Communication, Deoxyribonucleases metabolism, Epithelial Cells cytology, Extracellular Matrix metabolism, Human Umbilical Vein Endothelial Cells, Humans, Image Processing, Computer-Assisted, Oligonucleotides chemistry, Organoids cytology, Organoids physiology, Stromal Cells cytology, DNA chemistry, Extracellular Matrix chemistry, Tissue Engineering methods
- Abstract
Reconstituting tissues from their cellular building blocks facilitates the modeling of morphogenesis, homeostasis and disease in vitro. Here we describe DNA-programmed assembly of cells (DPAC), a method to reconstitute the multicellular organization of organoid-like tissues having programmed size, shape, composition and spatial heterogeneity. DPAC uses dissociated cells that are chemically functionalized with degradable oligonucleotide 'Velcro', allowing rapid, specific and reversible cell adhesion to other surfaces coated with complementary DNA sequences. DNA-patterned substrates function as removable and adhesive templates, and layer-by-layer DNA-programmed assembly builds arrays of tissues into the third dimension above the template. DNase releases completed arrays of organoid-like microtissues from the template concomitant with full embedding in a variety of extracellular matrix (ECM) gels. DPAC positions subpopulations of cells with single-cell spatial resolution and generates cultures several centimeters long. We used DPAC to explore the impact of ECM composition, heterotypic cell-cell interactions and patterns of signaling heterogeneity on collective cell behaviors.
- Published
- 2015
- Full Text
- View/download PDF
17. The senescent methylome and its relationship with cancer, ageing and germline genetic variation in humans.
- Author
-
Lowe R, Overhoff MG, Ramagopalan SV, Garbe JC, Koh J, Stampfer MR, Beach DH, Rakyan VK, and Bishop CL
- Subjects
- Adult, Cyclin-Dependent Kinase Inhibitor p16 genetics, Epigenesis, Genetic, Female, Genetic Variation, Humans, Polymorphism, Single Nucleotide, Young Adult, Aging genetics, Cellular Senescence genetics, DNA Methylation, Neoplasms genetics
- Abstract
Background: Cellular senescence is a stable arrest of proliferation and is considered a key component of processes associated with carcinogenesis and other ageing-related phenotypes. Here, we perform methylome analysis of actively dividing and deeply senescent normal human epithelial cells., Results: We identify senescence-associated differentially methylated positions (senDMPs) from multiple experiments using cells from one donor. We find that human senDMP epigenetic signatures are positively and significantly correlated with both cancer and ageing-associated methylation dynamics. We also identify germline genetic variants, including those associated with the p16INK4A locus, which are associated with the presence of in vivo senDMP signatures. Importantly, we also demonstrate that a single senDMP signature can be effectively reversed in a newly-developed protocol of transient senescence reversal., Conclusions: The senDMP signature has significant potential for understanding some of the key (epi)genetic etiological factors that may lead to cancer and age-related diseases in humans.
- Published
- 2015
- Full Text
- View/download PDF
18. Delineating transcriptional networks of prognostic gene signatures refines treatment recommendations for lymph node-negative breast cancer patients.
- Author
-
Lanigan F, Brien GL, Fan Y, Madden SF, Jerman E, Maratha A, Aloraifi F, Hokamp K, Dunne EJ, Lohan AJ, Flanagan L, Garbe JC, Stampfer MR, Fridberg M, Jirstrom K, Quinn CM, Loftus B, Gallagher WM, Geraghty J, and Bracken AP
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Breast Neoplasms metabolism, Breast Neoplasms therapy, Cell Proliferation genetics, Cells, Cultured, Cellular Senescence genetics, Cohort Studies, Female, Genes, p16, Humans, Lymphatic Metastasis genetics, Mammary Glands, Human cytology, Mammary Glands, Human metabolism, Mice, Middle Aged, Prognosis, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Receptors, Estrogen metabolism, Risk Factors, Tissue Array Analysis, Breast Neoplasms genetics, Gene Regulatory Networks
- Abstract
The majority of women diagnosed with lymph node-negative breast cancer are unnecessarily treated with damaging chemotherapeutics after surgical resection. This highlights the importance of understanding and more accurately predicting patient prognosis. In the present study, we define the transcriptional networks regulating well-established prognostic gene expression signatures. We find that the same set of transcriptional regulators consistently lie upstream of both 'prognosis' and 'proliferation' gene signatures, suggesting that a central transcriptional network underpins a shared phenotype within these signatures. Strikingly, the master transcriptional regulators within this network predict recurrence risk for lymph node-negative breast cancer better than currently used multigene prognostic assays, particularly in estrogen receptor-positive patients. Simultaneous examination of p16(INK4A) expression, which predicts tumours that have bypassed cellular senescence, revealed that intermediate levels of p16(INK4A) correlate with an intact pRB pathway and improved survival. A combination of these master transcriptional regulators and p16(INK4A), termed the OncoMasTR score, stratifies tumours based on their proliferative and senescence capacity, facilitating a clearer delineation of lymph node-negative breast cancer patients at high risk of recurrence, and thus requiring chemotherapy. Furthermore, OncoMasTR accurately classifies over 60% of patients as 'low risk', an improvement on existing prognostic assays, which has the potential to reduce overtreatment in early-stage patients. Taken together, the present study provides new insights into the transcriptional regulation of cellular proliferation in breast cancer and provides an opportunity to enhance and streamline methods of predicting breast cancer prognosis., (© 2015 FEBS.)
- Published
- 2015
- Full Text
- View/download PDF
19. Formation of spatially and geometrically controlled three-dimensional tissues in soft gels by sacrificial micromolding.
- Author
-
Cerchiari A, Garbe JC, Todhunter ME, Jee NY, Pinney JR, LaBarge MA, Desai TA, and Gartner ZJ
- Subjects
- Animals, Caco-2 Cells, Dogs, Humans, Madin Darby Canine Kidney Cells, Cell Culture Techniques methods, Hydrogels chemistry, Tissue Scaffolds chemistry
- Abstract
Patterned three-dimensional (3D) cell culture models aim to more accurately represent the in vivo architecture of a tissue for the purposes of testing drugs, studying multicellular biology, or engineering functional tissues. However, patterning 3D multicellular structures within very soft hydrogels (<500 Pa) that mimic the physicochemical environment of many tissues remains a challenge for existing methods. To overcome this challenge, we use a Sacrificial Micromolding technique to temporarily form spatially and geometrically defined 3D cell aggregates in degradable scaffolds before transferring and culturing them in a reconstituted extracellular matrix. Herein, we demonstrate that Sacrificial Micromolding (1) promotes cyst formation and proper polarization of established epithelial cell lines, (2) allows reconstitution of heterotypic cell-cell interactions in multicomponent epithelia, and (3) can be used to control the lumenization-state of epithelial cysts as a function of tissue size. In addition, we discuss the potential of Sacrificial Micromolding as a cell-patterning tool for future studies.
- Published
- 2015
- Full Text
- View/download PDF
20. Age and the means of bypassing stasis influence the intrinsic subtype of immortalized human mammary epithelial cells.
- Author
-
Lee JK, Garbe JC, Vrba L, Miyano M, Futscher BW, Stampfer MR, and LaBarge MA
- Abstract
Based on molecular features, breast cancers are grouped into intrinsic subtypes that have different prognoses and therapeutic response profiles. With increasing age, breast cancer incidence increases, with hormone receptor-positive and other luminal-like subtype tumors comprising a majority of cases. It is not known at what stage of tumor progression subtype specification occurs, nor how the process of aging affects the intrinsic subtype. We examined subtype markers in immortalized human mammary epithelial cell lines established following exposure of primary cultured cell strains to a two-step immortalization protocol that targets the two main barriers to immortality: stasis (stress-associated senescence) and replicative senescence. Cell lines derived from epithelial cells obtained from non-tumorous pre- and post-menopausal breast surgery tissues were compared. Additionally, comparisons were made between lines generated using two different genetic interventions to bypass stasis: transduction of either an shRNA that down-regulated p16(INK4A), or overexpressed constitutive active cyclin D1/CDK2. In all cases, the replicative senescence barrier was bypassed by transduction of c-Myc. Cells from all resulting immortal lines exhibited normal karyotypes. Immunofluorescence, flow cytometry, and gene expression analyses of lineage-specific markers were used to categorize the intrinsic subtypes of the immortalized lines. Bypassing stasis with p16 shRNA in young strains generated cell lines that were invariably basal-like, but the lines examined from older strains exhibited some luminal features such as keratin 19 and estrogen receptor expression. Overexpression of cyclin D1/CDK2 resulted in keratin 19 positive, luminal-like cell lines from both young and old strains, and the lines examined from older strains exhibited estrogen receptor expression. Thus age and the method of bypassing stasis independently influence the subtype of immortalized human mammary epithelial cells.
- Published
- 2015
- Full Text
- View/download PDF
21. A strategy for tissue self-organization that is robust to cellular heterogeneity and plasticity.
- Author
-
Cerchiari AE, Garbe JC, Jee NY, Todhunter ME, Broaders KE, Peehl DM, Desai TA, LaBarge MA, Thomson M, and Gartner ZJ
- Subjects
- Epithelial Cells cytology, Extracellular Matrix, Humans, Cell Communication, Mammary Glands, Human cytology
- Abstract
Developing tissues contain motile populations of cells that can self-organize into spatially ordered tissues based on differences in their interfacial surface energies. However, it is unclear how self-organization by this mechanism remains robust when interfacial energies become heterogeneous in either time or space. The ducts and acini of the human mammary gland are prototypical heterogeneous and dynamic tissues comprising two concentrically arranged cell types. To investigate the consequences of cellular heterogeneity and plasticity on cell positioning in the mammary gland, we reconstituted its self-organization from aggregates of primary cells in vitro. We find that self-organization is dominated by the interfacial energy of the tissue-ECM boundary, rather than by differential homo- and heterotypic energies of cell-cell interaction. Surprisingly, interactions with the tissue-ECM boundary are binary, in that only one cell type interacts appreciably with the boundary. Using mathematical modeling and cell-type-specific knockdown of key regulators of cell-cell cohesion, we show that this strategy of self-organization is robust to severe perturbations affecting cell-cell contact formation. We also find that this mechanism of self-organization is conserved in the human prostate. Therefore, a binary interfacial interaction with the tissue boundary provides a flexible and generalizable strategy for forming and maintaining the structure of two-component tissues that exhibit abundant heterogeneity and plasticity. Our model also predicts that mutations affecting binary cell-ECM interactions are catastrophic and could contribute to loss of tissue architecture in diseases such as breast cancer.
- Published
- 2015
- Full Text
- View/download PDF
22. A lincRNA connected to cell mortality and epigenetically-silenced in most common human cancers.
- Author
-
Vrba L, Garbe JC, Stampfer MR, and Futscher BW
- Subjects
- Aged, 80 and over, Cell Line, Tumor, Cell Survival, Epigenesis, Genetic, Epithelial Cells, Female, Gene Expression Profiling methods, Humans, Middle Aged, Oligonucleotide Array Sequence Analysis methods, Young Adult, DNA Methylation, Mammary Glands, Human cytology, Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
Immortality is an essential characteristic of human carcinoma cells. We recently developed an efficient, reproducible method that immortalizes human mammary epithelial cells (HMEC) in the absence of gross genomic changes by targeting 2 critical senescence barriers. Consistent transcriptomic changes associated with immortality were identified using microarray analysis of isogenic normal finite pre-stasis, abnormal finite post-stasis, and immortal HMECs from 4 individuals. A total of 277 genes consistently changed in cells that transitioned from post-stasis to immortal. Gene ontology analysis of affected genes revealed biological processes significantly altered in the immortalization process. These immortalization-associated changes showed striking similarity to the gene expression changes seen in The Cancer Genome Atlas (TCGA) clinical breast cancer data. The most dramatic change in gene expression seen during the immortalization step was the downregulation of an unnamed, incompletely annotated transcript that we called MORT, for mortality, since its expression was closely associated with the mortal, finite lifespan phenotype. We show here that MORT (ZNF667-AS1) is expressed in all normal finite lifespan human cells examined to date and is lost in immortalized HMEC. MORT gene silencing at the mortal/immortal boundary was due to DNA hypermethylation of its CpG island promoter. This epigenetic silencing is also seen in human breast cancer cell lines and in a majority of human breast tumor tissues. The functional importance of DNA hypermethylation in MORT gene silencing is supported by the ability of 5-aza-2'-deoxycytidine to reactivate MORT expression. Analysis of TCGA data revealed deregulation of MORT expression due to DNA hypermethylation in 15 out of the 17 most common human cancers. The epigenetic silencing of MORT in a large majority of the common human cancers suggests a potential fundamental role in cellular immortalization during human carcinogenesis.
- Published
- 2015
- Full Text
- View/download PDF
23. Age-related dysfunction in mechanotransduction impairs differentiation of human mammary epithelial progenitors.
- Author
-
Pelissier FA, Garbe JC, Ananthanarayanan B, Miyano M, Lin C, Jokela T, Kumar S, Stampfer MR, Lorens JB, and LaBarge MA
- Subjects
- Acyltransferases, Adaptor Proteins, Signal Transducing metabolism, Adult, Age Factors, Cell Differentiation physiology, Epithelial Cells cytology, Female, Humans, Mammary Glands, Human metabolism, Mechanotransduction, Cellular physiology, Middle Aged, Phosphoproteins metabolism, Stem Cells metabolism, Transcription Factors metabolism, YAP-Signaling Proteins, Epithelial Cells metabolism, Mammary Glands, Human cytology, Stem Cells cytology
- Abstract
Dysfunctional progenitor and luminal cells with acquired basal cell properties accumulate during human mammary epithelial aging for reasons not understood. Multipotent progenitors from women aged <30 years were exposed to a physiologically relevant range of matrix elastic modulus (stiffness). Increased stiffness causes a differentiation bias towards myoepithelial cells while reducing production of luminal cells and progenitor maintenance. Lineage representation in progenitors from women >55 years is unaffected by physiological stiffness changes. Efficient activation of Hippo pathway transducers YAP and TAZ is required for the modulus-dependent myoepithelial/basal bias in younger progenitors. In older progenitors, YAP and TAZ are activated only when stressed with extraphysiologically stiff matrices, which bias differentiation towards luminal-like phenotypes. In vivo YAP is primarily active in myoepithelia of younger breasts, but localization and activity increases in luminal cells with age. Thus, aging phenotypes of mammary epithelia may arise partly because alterations in Hippo pathway activation impair microenvironment-directed differentiation and lineage specificity., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
24. Cellular senescence mediated by p16INK4A-coupled miRNA pathways.
- Author
-
Overhoff MG, Garbe JC, Koh J, Stampfer MR, Beach DH, and Bishop CL
- Subjects
- Cells, Cultured, Feedback, Physiological, Fibroblasts cytology, Fibroblasts metabolism, Gene Silencing, Humans, Polycomb-Group Proteins genetics, Polycomb-Group Proteins metabolism, Young Adult, Cellular Senescence genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Epigenesis, Genetic, MicroRNAs metabolism
- Abstract
p16 is a key regulator of cellular senescence, yet the drivers of this stable state of proliferative arrest are not well understood. Here, we identify 22 senescence-associated microRNAs (SA-miRNAs) in normal human mammary epithelial cells. We show that SA-miRNAs-26b, 181a, 210 and 424 function in concert to directly repress expression of Polycomb group (PcG) proteins CBX7, embryonic ectoderm development (EED), enhancer of zeste homologue 2 (EZH2) and suppressor of zeste 12 homologue (Suz12), thereby activating p16. We demonstrate the existence of a tight positive feedback loop in which SA-miRNAs activate and re-enforce the expression of other SA-miRNA members. In contrast, PcG members restrain senescence by epigenetically repressing the expression of these SA-miRNAs. Importantly, loss of p16 leads to repression of SA-miRNA expression, intimately coupling this effector of senescence to the SA-miRNA/PcG self-regulatory loop. Taken together, our findings illuminate an important regulatory axis that underpins the transition from proliferation to cellular senescence.
- Published
- 2014
- Full Text
- View/download PDF
25. Immortalization of normal human mammary epithelial cells in two steps by direct targeting of senescence barriers does not require gross genomic alterations.
- Author
-
Garbe JC, Vrba L, Sputova K, Fuchs L, Novak P, Brothman AR, Jackson M, Chin K, LaBarge MA, Watts G, Futscher BW, and Stampfer MR
- Subjects
- Cells, Cultured, Cellular Senescence, Chromosome Aberrations, Cyclin-Dependent Kinase Inhibitor p16 antagonists & inhibitors, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Epigenesis, Genetic, Genomic Instability, Histones metabolism, Humans, Karyotyping, Mammary Glands, Human metabolism, Promoter Regions, Genetic, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, RNA, Small Interfering metabolism, Telomerase genetics, Telomerase metabolism, Mammary Glands, Human cytology
- Abstract
Telomerase reactivation and immortalization are critical for human carcinoma progression. However, little is known about the mechanisms controlling this crucial step, due in part to the paucity of experimentally tractable model systems that can examine human epithelial cell immortalization as it might occur in vivo. We achieved efficient non-clonal immortalization of normal human mammary epithelial cells (HMEC) by directly targeting the 2 main senescence barriers encountered by cultured HMEC. The stress-associated stasis barrier was bypassed using shRNA to p16(INK4); replicative senescence due to critically shortened telomeres was bypassed in post-stasis HMEC by c-MYC transduction. Thus, 2 pathologically relevant oncogenic agents are sufficient to immortally transform normal HMEC. The resultant non-clonal immortalized lines exhibited normal karyotypes. Most human carcinomas contain genomically unstable cells, with widespread instability first observed in vivo in pre-malignant stages; in vitro, instability is seen as finite cells with critically shortened telomeres approach replicative senescence. Our results support our hypotheses that: (1) telomere-dysfunction induced genomic instability in pre-malignant finite cells may generate the errors required for telomerase reactivation and immortalization, as well as many additional "passenger" errors carried forward into resulting carcinomas; (2) genomic instability during cancer progression is needed to generate errors that overcome tumor suppressive barriers, but not required per se; bypassing the senescence barriers by direct targeting eliminated a need for genomic errors to generate immortalization. Achieving efficient HMEC immortalization, in the absence of "passenger" genomic errors, should facilitate examination of telomerase regulation during human carcinoma progression, and exploration of agents that could prevent immortalization.
- Published
- 2014
- Full Text
- View/download PDF
26. Aging phenotypes in cultured normal human mammary epithelial cells are correlated with decreased telomerase activity independent of telomere length.
- Author
-
Sputova K, Garbe JC, Pelissier FA, Chang E, Stampfer MR, and LaBarge MA
- Abstract
Background: Shortening of telomeres, which are essential for maintenance of genomic integrity, is a mechanism commonly associated with the aging process. Here we ascertained whether changes in telomere lengths or telomerase activity correlated with age in normal human mammary epithelial cells (HMEC), or with phenotypes of aging in breast. Accordingly, flow cytometry fluorescence in situ hybridization (flowFISH) was used to determine relative telomere lengths (RTL), and telomerase activity was measured by the telomeric repeat amplification protocol (TRAP), in a collection of 41 primary HMEC strains established from women aged 16 to 91 years., Results: RTL measurements of HMEC strains that were heterogeneous with respect to lineage composition revealed no significant associations between telomere length with age, maximum observed population doublings, or with lineage composition of the strains. However, within strains, luminal epithelial and cKit-expressing epithelial progenitor cells that were flow cytometry-enriched from individual HMEC strains exhibited significantly shorter telomeres relative to isogenic myoepithelial cells (P < 0.01). In unsorted strains, detectable telomerase activity did not correlate with RTL. Telomerase activity declined with age; the average age of strains that exhibited TRAP activity was 29.7 ± 3.9y, whereas the average age of strains with no detectable TRAP activity was 49.0 ± 4.9y (P < 0.01). Non-detectable TRAP activity also was correlated with phenotypes of aging previously described in HMEC strains; increased proportions of CD227-expressing luminal epithelial cells (P < 0.05) and cKit-expressing progenitor cells (P < 0.05)., Conclusions: Telomere shortening did not correlate with the chronological ages of HMEC strains, whereas decreased telomerase activity correlated with age and with lineage distribution phenotypes characteristic of aging.
- Published
- 2013
- Full Text
- View/download PDF
27. Processing of human reduction mammoplasty and mastectomy tissues for cell culture.
- Author
-
Labarge MA, Garbe JC, and Stampfer MR
- Subjects
- Epithelial Cells cytology, Female, Humans, Mammaplasty, Mastectomy, Cytological Techniques methods, Mammary Glands, Human cytology, Mammary Glands, Human surgery
- Abstract
Experimental examination of normal human mammary epithelial cell (HMEC) behavior, and how normal cells acquire abnormal properties, can be facilitated by in vitro culture systems that more accurately model in vivo biology. The use of human derived material for studying cellular differentiation, aging, senescence, and immortalization is particularly advantageous given the many significant molecular differences in these properties between human and commonly utilized rodent cells. Mammary cells present a convenient model system because large quantities of normal and abnormal tissues are available due to the frequency of reduction mammoplasty and mastectomy surgeries. The mammary gland consists of a complex admixture of many distinct cell types, e.g., epithelial, adipose, mesenchymal, endothelial. The epithelial cells are responsible for the differentiated mammary function of lactation, and are also the origin of the vast majority of human breast cancers. We have developed methods to process mammary gland surgical discard tissues into pure epithelial components as well as mesenchymal cells. The processed material can be stored frozen indefinitely, or initiated into primary culture. Surgical discard material is transported to the laboratory and manually dissected to enrich for epithelial containing tissue. Subsequent digestion of the dissected tissue using collagenase and hyaluronidase strips stromal material from the epithelia at the basement membrane. The resulting small pieces of the epithelial tree (organoids) can be separated from the digested stroma by sequential filtration on membranes of fixed pore size. Depending upon pore size, fractions can be obtained consisting of larger ductal/alveolar pieces, smaller alveolar clusters, or stromal cells. We have observed superior growth when cultures are initiated as organoids rather than as dissociated single cells. Placement of organoids in culture using low-stress inducing media supports long-term growth of normal HMEC with markers of multiple lineage types (myoepithelial, luminal, progenitor). Sufficient numbers of cells can be obtained from one individual's tissue to allow extensive experimental examination using standardized cell batches, as well as interrogation using high throughput modalities. Cultured HMEC have been employed in a wide variety of studies examining the normal processes governing growth, differentiation, aging, and senescence, and how these normal processes are altered during immortal and malignant transformation. The effects of growth in the presence of extracellular matrix material, other cell types, and/or 3D culture can be compared with growth on plastic. Cultured HMEC, starting with normal cells, provide an experimentally tractable system to examine factors that may propel or prevent human aging and carcinogenesis.
- Published
- 2013
- Full Text
- View/download PDF
28. Accumulation of multipotent progenitors with a basal differentiation bias during aging of human mammary epithelia.
- Author
-
Garbe JC, Pepin F, Pelissier FA, Sputova K, Fridriksdottir AJ, Guo DE, Villadsen R, Park M, Petersen OW, Borowsky AD, Stampfer MR, and Labarge MA
- Subjects
- Aged, Aged, 80 and over, Cells, Cultured, Epithelial Cells cytology, Female, Humans, Middle Aged, Phenotype, Aging, Cell Differentiation, Cellular Senescence, Mammary Glands, Human cytology, Multipotent Stem Cells physiology
- Abstract
Women older than 50 years account for 75% of new breast cancer diagnoses, and the majority of these tumors are of a luminal subtype. Although age-associated changes, including endocrine profiles and alterations within the breast microenvironment, increase cancer risk, an understanding of the cellular and molecular mechanisms that underlies these observations is lacking. In this study, we generated a large collection of normal human mammary epithelial cell strains from women ages 16 to 91 years, derived from primary tissues, to investigate the molecular changes that occur in aging breast cells. We found that in finite lifespan cultured and uncultured epithelial cells, aging is associated with a reduction of myoepithelial cells and an increase in luminal cells that express keratin 14 and integrin-α6, a phenotype that is usually expressed exclusively in myoepithelial cells in women younger than 30 years. Changes to the luminal lineage resulted from age-dependent expansion of defective multipotent progenitors that gave rise to incompletely differentiated luminal or myoepithelial cells. The aging process therefore results in both a shift in the balance of luminal/myoepithelial lineages and to changes in the functional spectrum of multipotent progenitors, which together increase the potential for malignant transformation. Together, our findings provide a cellular basis to explain the observed vulnerability to breast cancer that increases with age.
- Published
- 2012
- Full Text
- View/download PDF
29. Cell-type specific DNA methylation patterns define human breast cellular identity.
- Author
-
Novak P, Stampfer MR, Munoz-Rodriguez JL, Garbe JC, Ehrich M, Futscher BW, and Jensen TJ
- Subjects
- Female, Histones metabolism, Humans, Oligonucleotide Array Sequence Analysis, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Breast cytology, Breast metabolism, DNA Methylation genetics
- Abstract
DNA methylation plays a role in a variety of biological processes including embryonic development, imprinting, X-chromosome inactivation, and stem cell differentiation. Tissue specific differential methylation has also been well characterized. We sought to extend these studies to create a map of differential DNA methylation between different cell types derived from a single tissue. Using three pairs of isogenic human mammary epithelial and fibroblast cells, promoter region DNA methylation was characterized using MeDIP coupled to microarray analysis. Comparison of DNA methylation between these cell types revealed nearly three thousand cell-type specific differentially methylated regions (ctDMRs). MassARRAY was performed upon 87 ctDMRs to confirm and quantify differential DNA methylation. Each of the examined regions exhibited statistically significant differences ranging from 10-70%. Gene ontology analysis revealed the overrepresentation of many transcription factors involved in developmental processes. Additionally, we have shown that ctDMRs are associated with histone related epigenetic marks and are often aberrantly methylated in breast cancer. Overall, our data suggest that there are thousands of ctDMRs which consistently exhibit differential DNA methylation and may underlie cell type specificity in human breast tissue. In addition, we describe the pathways affected by these differences and provide insight into the molecular mechanisms and physiological overlap between normal cellular differentiation and breast carcinogenesis.
- Published
- 2012
- Full Text
- View/download PDF
30. Epigenetic regulation of normal human mammary cell type-specific miRNAs.
- Author
-
Vrba L, Garbe JC, Stampfer MR, and Futscher BW
- Subjects
- Breast cytology, Cell Line, Female, Fibroblasts cytology, Humans, Organ Specificity physiology, Breast metabolism, Epigenesis, Genetic physiology, Fibroblasts metabolism, MicroRNAs biosynthesis
- Abstract
Epigenetic mechanisms are important regulators of cell type-specific genes, including miRNAs. In order to identify cell type-specific miRNAs regulated by epigenetic mechanisms, we undertook a global analysis of miRNA expression and epigenetic states in three isogenic pairs of human mammary epithelial cells (HMEC) and human mammary fibroblasts (HMF), which represent two differentiated cell types typically present within a given organ, each with a distinct phenotype and a distinct epigenotype. While miRNA expression and epigenetic states showed strong interindividual concordance within a given cell type, almost 10% of the expressed miRNA showed a cell type-specific pattern of expression that was linked to the epigenetic state of their promoter. The tissue-specific miRNA genes were epigenetically repressed in nonexpressing cells by DNA methylation (38%) and H3K27me3 (58%), with only a small set of miRNAs (21%) showing a dual epigenetic repression where both DNA methylation and H3K27me3 were present at their promoters, such as MIR10A and MIR10B. Individual miRNA clusters of closely related miRNA gene families can each display cell type-specific repression by the same or complementary epigenetic mechanisms, such as the MIR200 family, and MIR205, where fibroblasts repress MIR200C/141 by DNA methylation, MIR200A/200B/429 by H3K27me3, and MIR205 by both DNA methylation and H3K27me3. Since deregulation of many of the epigenetically regulated miRNAs that we identified have been linked to disease processes such as cancer, it is predicted that compromise of the epigenetic control mechanisms is important for this process. Overall, these results highlight the importance of epigenetic regulation in the control of normal cell type-specific miRNA expression.
- Published
- 2011
- Full Text
- View/download PDF
31. Self-organization is a dynamic and lineage-intrinsic property of mammary epithelial cells.
- Author
-
Chanson L, Brownfield D, Garbe JC, Kuhn I, Stampfer MR, Bissell MJ, and LaBarge MA
- Subjects
- Actomyosin physiology, Adherens Junctions physiology, Body Patterning, Cadherins physiology, Cell Lineage, Cells, Cultured, Coculture Techniques, Humans, Epithelial Cells cytology, Mammary Glands, Human cytology
- Abstract
Loss of organization is a principle feature of cancers; therefore it is important to understand how normal adult multilineage tissues, such as bilayered secretory epithelia, establish and maintain their architectures. The self-organization process that drives heterogeneous mixtures of cells to form organized tissues is well studied in embryology and with mammalian cell lines that were abnormal or engineered. Here we used a micropatterning approach that confined cells to a cylindrical geometry combined with an algorithm to quantify changes of cellular distribution over time to measure the ability of different cell types to self-organize relative to each other. Using normal human mammary epithelial cells enriched into pools of the two principal lineages, luminal and myoepithelial cells, we demonstrated that bilayered organization in mammary epithelium was driven mainly by lineage-specific differential E-cadherin expression, but that P-cadherin contributed specifically to organization of the myoepithelial layer. Disruption of the actomyosin network or of adherens junction proteins resulted in either prevention of bilayer formation or loss of preformed bilayers, consistent with continual sampling of the local microenvironment by cadherins. Together these data show that self-organization is an innate and reversible property of communities of normal adult human mammary epithelial cells.
- Published
- 2011
- Full Text
- View/download PDF
32. Primary cilium-dependent and -independent Hedgehog signaling inhibits p16(INK4A).
- Author
-
Bishop CL, Bergin AM, Fessart D, Borgdorff V, Hatzimasoura E, Garbe JC, Stampfer MR, Koh J, and Beach DH
- Subjects
- Female, Genome, Human genetics, Humans, Protein Interaction Mapping, RNA, Small Interfering metabolism, Young Adult, Zinc Finger Protein Gli2, Cilia metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Hedgehog Proteins metabolism, Kruppel-Like Transcription Factors metabolism, Nuclear Proteins metabolism, Signal Transduction
- Abstract
In a genome-wide siRNA analysis of p16(INK4a) (p16) modulators, we identify the Hedgehog (Hh) pathway component SUFU and formally demonstrate that Hh signaling promotes mitogenesis by suppression of p16. A fragment of the Hh-responsive GLI2 transcription factor directly binds and inhibits the p16 promoter and senescence is associated with the loss of nuclear GLI2. Hh components partially reside in the primary cilium (PC), and the small fraction of cells in mass culture that elaborate a PC have the lowest expression of p16. Suppression of p16 is effected by both PC-dependent and -independent routes, and ablation of p16 renders cells insensitive to an Hh inhibitor and increases PC formation. These results directly link a well-established developmental mitogenic pathway with a key tumor suppressor and contribute to the molecular understanding of replicative senescence, Hh-mediated oncogenesis, and potentially the role of p16 in aging., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
33. Role for DNA methylation in the regulation of miR-200c and miR-141 expression in normal and cancer cells.
- Author
-
Vrba L, Jensen TJ, Garbe JC, Heimark RL, Cress AE, Dickinson S, Stampfer MR, and Futscher BW
- Subjects
- Cell Line, Cell Line, Tumor, Chromatin Immunoprecipitation, CpG Islands, Histones metabolism, Humans, Polymerase Chain Reaction, DNA Methylation, Gene Expression Regulation, MicroRNAs genetics
- Abstract
Background: The microRNA-200 family participates in the maintenance of an epithelial phenotype and loss of its expression can result in epithelial to mesenchymal transition (EMT). Furthermore, the loss of expression of miR-200 family members is linked to an aggressive cancer phenotype. Regulation of the miR-200 family expression in normal and cancer cells is not fully understood., Methodology/principal Findings: Epigenetic mechanisms participate in the control of miR-200c and miR-141 expression in both normal and cancer cells. A CpG island near the predicted mir-200c/mir-141 transcription start site shows a striking correlation between miR-200c and miR-141 expression and DNA methylation in both normal and cancer cells, as determined by MassARRAY technology. The CpG island is unmethylated in human miR-200/miR-141 expressing epithelial cells and in miR-200c/miR-141 positive tumor cells. The CpG island is heavily methylated in human miR-200c/miR-141 negative fibroblasts and miR-200c/miR-141 negative tumor cells. Mouse cells show a similar inverse correlation between DNA methylation and miR-200c expression. Enrichment of permissive histone modifications, H3 acetylation and H3K4 trimethylation, is seen in normal miR-200c/miR-141-positive epithelial cells, as determined by chromatin immunoprecipitation coupled to real-time PCR. In contrast, repressive H3K9 dimethylation marks are present in normal miR-200c/miR-141-negative fibroblasts and miR-200c/miR-141 negative cancer cells and the permissive histone modifications are absent. The epigenetic modifier drug, 5-aza-2'-deoxycytidine, reactivates miR-200c/miR-141 expression showing that epigenetic mechanisms play a functional role in their transcriptional control., Conclusions/significance: We report that DNA methylation plays a role in the normal cell type-specific expression of miR-200c and miR-141 and this role appears evolutionarily conserved, since similar results were obtained in mouse. Aberrant DNA methylation of the miR-200c/141 CpG island is closely linked to their inappropriate silencing in cancer cells. Since the miR-200c cluster plays a significant role in EMT, our results suggest an important role for DNA methylation in the control of phenotypic conversions in normal cells.
- Published
- 2010
- Full Text
- View/download PDF
34. Molecular distinctions between stasis and telomere attrition senescence barriers shown by long-term culture of normal human mammary epithelial cells.
- Author
-
Garbe JC, Bhattacharya S, Merchant B, Bassett E, Swisshelm K, Feiler HS, Wyrobek AJ, and Stampfer MR
- Subjects
- Adolescent, Adult, Cell Growth Processes drug effects, Cell Growth Processes physiology, Cells, Cultured, Culture Media, DNA Damage, Epithelial Cells cytology, Epithelial Cells drug effects, Epithelial Cells metabolism, Epithelial Cells ultrastructure, Female, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts ultrastructure, Gene Expression, Humans, Mammary Glands, Human cytology, Mammary Glands, Human drug effects, Mammary Glands, Human metabolism, Oxytocin pharmacology, Protein Biosynthesis, Telomere genetics, Transcription, Genetic, Young Adult, Mammary Glands, Human ultrastructure, Telomere metabolism
- Abstract
Normal human epithelial cells in culture have generally shown a limited proliferative potential of approximately 10 to 40 population doublings before encountering a stress-associated senescence barrier (stasis) associated with elevated levels of cyclin-dependent kinase inhibitors p16 and/or p21. We now show that simple changes in medium composition can expand the proliferative potential of human mammary epithelial cells (HMEC) initiated as primary cultures to 50 to 60 population doublings followed by p16-positive, senescence-associated beta-galactosidase-positive stasis. We compared the properties of growing and senescent pre-stasis HMEC with growing and senescent post-selection HMEC, that is, cells grown in a serum-free medium that overcame stasis via silencing of p16 expression and that display senescence associated with telomere dysfunction. Cultured pre-stasis populations contained cells expressing markers associated with luminal and myoepithelial HMEC lineages in vivo in contrast to the basal-like phenotype of the post-selection HMEC. Gene transcript and protein expression, DNA damage-associated markers, mean telomere restriction fragment length, and genomic stability differed significantly between HMEC populations at the stasis versus telomere dysfunction senescence barriers. Senescent isogenic fibroblasts showed greater similarity to HMEC at stasis than at telomere dysfunction, although their gene transcript profile was distinct from HMEC at both senescence barriers. These studies support our model of the senescence barriers encountered by cultured HMEC in which the first barrier, stasis, is retinoblastoma-mediated and independent of telomere length, whereas a second barrier (agonescence or crisis) results from telomere attrition leading to telomere dysfunction. Additionally, the ability to maintain long-term growth of genomically stable multilineage pre-stasis HMEC populations can greatly enhance experimentation with normal HMEC.
- Published
- 2009
- Full Text
- View/download PDF
35. Stepwise DNA methylation changes are linked to escape from defined proliferation barriers and mammary epithelial cell immortalization.
- Author
-
Novak P, Jensen TJ, Garbe JC, Stampfer MR, and Futscher BW
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Humans, Immunoprecipitation, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic, Cell Proliferation, DNA Methylation, Mammary Glands, Human cytology
- Abstract
The timing and progression of DNA methylation changes during carcinogenesis are not completely understood. To develop a timeline of aberrant DNA methylation events during malignant transformation, we analyzed genome-wide DNA methylation patterns in an isogenic human mammary epithelial cell (HMEC) culture model of transformation. To acquire immortality and malignancy, the cultured finite lifespan HMEC must overcome two distinct proliferation barriers. The first barrier, stasis, is mediated by the retinoblastoma protein and can be overcome by loss of p16(INK4A) expression. HMEC that escape stasis and continue to proliferate become genomically unstable before encountering a second more stringent proliferation barrier, telomere dysfunction due to telomere attrition. Rare cells that acquire telomerase expression may escape this barrier, become immortal, and develop further malignant properties. Our analysis of HMEC transitioning from finite lifespan to malignantly transformed showed that aberrant DNA methylation changes occur in a stepwise fashion early in the transformation process. The first aberrant DNA methylation step coincides with overcoming stasis, and results in few to hundreds of changes, depending on how stasis was overcome. A second step coincides with immortalization and results in hundreds of additional DNA methylation changes regardless of the immortalization pathway. A majority of these DNA methylation changes are also found in malignant breast cancer cells. These results show that large-scale epigenetic remodeling occurs in the earliest steps of mammary carcinogenesis, temporally links DNA methylation changes and overcoming cellular proliferation barriers, and provides a bank of potential epigenetic biomarkers that may prove useful in breast cancer risk assessment.
- Published
- 2009
- Full Text
- View/download PDF
36. Inactivation of p53 function in cultured human mammary epithelial cells turns the telomere-length dependent senescence barrier from agonescence into crisis.
- Author
-
Garbe JC, Holst CR, Bassett E, Tlsty T, and Stampfer MR
- Subjects
- Cell Death, Cell Line, Cell Proliferation, Cell Shape, DNA Damage genetics, Humans, Tumor Suppressor Protein p53 genetics, Cellular Senescence physiology, Epithelial Cells cytology, Epithelial Cells metabolism, Mammary Glands, Human cytology, Telomere genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Cultured human mammary epithelial cells (HMEC) encounter two distinct barriers to indefinite growth. The first barrier, originally termed selection, can be overcome through loss of expression of the cyclin-dependent kinase inhibitor p16(INK4A). The resultant p16-, p53+ post-selection HMEC encounter a second barrier, termed agonescence, associated with critically shortened telomeres and widespread chromosomal aberrations. Although some cell death is present at agonescence, the majority of the population retains long-term viability. We now show that abrogation of p53 function in post-selection HMEC inactivates cell cycle checkpoints and changes the mostly viable agonescence barrier into a crisis-like barrier with massive cell death. In contrast, inactivation of p53 does not affect the ability of HMEC to overcome the first barrier. These data indicate that agonescence and crisis represent two different forms of a telomere-length dependent proliferation barrier. Altogether, our data suggest a modified model of HMEC senescence barriers. We propose that the first barrier is Rb-mediated and largely or completely independent of telomere length. This barrier is now being termed stasis, for stress-associated senescence. The second barrier (agonescence or crisis) results from ongoing telomere erosion leading to critically short telomeres and telomere dysfunction.
- Published
- 2007
- Full Text
- View/download PDF
37. Transcriptional changes associated with breast cancer occur as normal human mammary epithelial cells overcome senescence barriers and become immortalized.
- Author
-
Li Y, Pan J, Li JL, Lee JH, Tunkey C, Saraf K, Garbe JC, Whitley MZ, Jelinsky SA, Stampfer MR, and Haney SA
- Subjects
- Cell Nucleus metabolism, Cluster Analysis, Epithelial Cells physiology, Gene Expression Profiling, Humans, Mammary Glands, Human cytology, Polymerase Chain Reaction methods, Promoter Regions, Genetic, Receptor, ErbB-2 metabolism, Regulatory Elements, Transcriptional, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Breast Neoplasms genetics, Epithelial Cells metabolism, Gene Expression Regulation, Neoplastic, Mammary Glands, Human metabolism, Transcription, Genetic
- Abstract
Background: Human mammary epithelial cells (HMEC) overcome two well-characterized genetic and epigenetic barriers as they progress from primary cells to fully immortalized cell lines in vitro. Finite lifespan HMEC overcome an Rb-mediated stress-associated senescence barrier (stasis), and a stringent, telomere-length dependent, barrier (agonescence or crisis, depending on p53 status). HMEC that have overcome the second senescence barrier are immortalized., Methods: We have characterized pre-stasis, post-selection (post-stasis, with p16 silenced), and fully immortalized HMEC by transcription profiling and RT-PCR. Four pre-stasis and seven post-selection HMEC samples, along with 10 representatives of fully immortalized breast epithelial cell lines, were profiled using Affymetrix U133A/B chips and compared using both supervised and unsupervised clustering. Datasets were validated by RT-PCR for a select set of genes. Quantitative immunofluorescence was used to assess changes in transcriptional regulators associated with the gene expression changes., Results: The most dramatic and uniform changes we observed were in a set of about 30 genes that are characterized as a "cancer proliferation cluster," which includes genes expressed during mitosis (CDC2, CDC25, MCM2, PLK1) and following DNA damage. The increased expression of these genes was particularly concordant in the fully immortalized lines. Additional changes were observed in IFN-regulated genes in some post-selection and fully immortalized cultures. Nuclear localization was observed for several transcriptional regulators associated with expression of these genes in post-selection and immortalized HMEC, including Rb, Myc, BRCA1, HDAC3 and SP1., Conclusion: Gene expression profiles and cytological changes in related transcriptional regulators indicate that immortalized HMEC resemble non-invasive breast cancers, such as ductal and lobular carcinomas in situ, and are strikingly distinct from finite-lifespan HMEC, particularly with regard to genes involved in proliferation, cell cycle regulation, chromosome structure and the DNA damage response. The comparison of HMEC profiles with lines harboring oncogenic changes (e.g. overexpression of Her-2neu, loss of p53 expression) identifies genes involved in tissue remodeling as well as proinflamatory cytokines and S100 proteins. Studies on carcinogenesis using immortalized cell lines as starting points or "normal" controls need to account for the significant pre-existing genetic and epigenetic changes inherent in such lines before results can be broadly interpreted.
- Published
- 2007
- Full Text
- View/download PDF
38. Caspase-independent cytochrome c release is a sensitive measure of low-level apoptosis in cell culture models.
- Author
-
Goldstein JC, Rodier F, Garbe JC, Stampfer MR, and Campisi J
- Subjects
- Animals, Caspase Inhibitors, Cells, Cultured, DNA Damage, DNA Fragmentation, Fibroblasts cytology, Fibroblasts enzymology, Fibroblasts metabolism, Humans, Mammary Glands, Human cytology, Mammary Glands, Human enzymology, Mammary Glands, Human metabolism, Mice, Apoptosis physiology, Caspases metabolism, Cytochromes c metabolism
- Abstract
Age-associated loss of tissue function and several chronic diseases may derive in part from the cumulative effects of subtle changes in the level of apoptotic cell death. Because apoptosis is rapid and undetectable once complete, small changes in its incidence are difficult to detect, even in well-controlled cell cultures. We describe a new apoptosis assay that provides greater sensitivity than conventional assays because it measures the accumulation of apoptotic cells. Human and mouse fibroblasts and human mammary epithelial cells that initiated apoptosis were preserved for 3 days by inhibiting caspase activity using the chemical inhibitor Q-VD-OPH (QVD). Cells suspended in the process of apoptosis were scored by immunostaining for cytochrome c, which redistributed from mitochondria in healthy cells to the cytoplasm in dying cells. This caspase-independent cytochrome c release (CICR) assay was more sensitive than several conventional assays when apoptosis was induced by actinomycin D, and detected cumulative background levels of apoptosis over a 3-day interval. Using this assay, we show that normal fibroblasts undergo very little apoptosis upon X-irradiation, indicating dominance of the senescence response in this cell type. Further, apoptosis increased subtly but measurably when human mammary epithelial and skin fibroblast cells entered crisis, indicating that cell death during crisis is largely non-apoptotic.
- Published
- 2005
- Full Text
- View/download PDF
39. Chromatin inactivation precedes de novo DNA methylation during the progressive epigenetic silencing of the RASSF1A promoter.
- Author
-
Strunnikova M, Schagdarsurengin U, Kehlen A, Garbe JC, Stampfer MR, and Dammann R
- Subjects
- Cell Line, Tumor, Cellular Senescence genetics, CpG Islands genetics, DNA genetics, DNA metabolism, Humans, Regulatory Sequences, Nucleic Acid genetics, Sp1 Transcription Factor metabolism, Sulfites metabolism, Chromatin genetics, Chromatin metabolism, DNA Methylation, Gene Silencing, Promoter Regions, Genetic genetics, Tumor Suppressor Proteins genetics
- Abstract
Epigenetic inactivation of the RASSF1A tumor suppressor by CpG island methylation was frequently detected in cancer. However, the mechanisms of this aberrant DNA methylation are unknown. In the RASSF1A promoter, we characterized four Sp1 sites, which are frequently methylated in cancer. We examined the functional relationship between DNA methylation, histone modification, Sp1 binding, and RASSF1A expression in proliferating human mammary epithelial cells. With increasing passages, the transcription of RASSF1A was dramatically silenced. This inactivation was associated with deacetylation and lysine 9 trimethylation of histone H3 and an impaired binding of Sp1 at the RASSF1A promoter. In mammary epithelial cells that had overcome a stress-associated senescence barrier, a spreading of DNA methylation in the CpG island promoter was observed. When the RASSF1A-silenced cells were treated with inhibitors of DNA methyltransferase and histone deacetylase, binding of Sp1 and expression of RASSF1A reoccurred. In summary, we observed that histone H3 deacetylation and H3 lysine 9 trimethylation occur in the same time window as gene inactivation and precede DNA methylation. Our data suggest that in epithelial cells, histone inactivation may trigger de novo DNA methylation of the RASSF1A promoter and this system may serve as a model for CpG island inactivation of tumor suppressor genes.
- Published
- 2005
- Full Text
- View/download PDF
40. p57KIP2 expression and loss of heterozygosity during immortal conversion of cultured human mammary epithelial cells.
- Author
-
Nijjar T, Wigington D, Garbe JC, Waha A, Stampfer MR, and Yaswen P
- Subjects
- Alleles, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p57, DNA-Binding Proteins, Epithelial Cells chemistry, Epithelial Cells pathology, Female, Humans, Nuclear Proteins genetics, Nuclear Proteins physiology, RNA, Messenger analysis, Telomerase genetics, Telomere, Breast chemistry, Breast pathology, Cell Transformation, Neoplastic, Enzyme Inhibitors analysis, Loss of Heterozygosity, Nuclear Proteins analysis, RNA
- Abstract
We have uncovered a novel role for the cyclin-dependent kinase inhibitor, p57KIP2, during the immortalization of cultured human mammary epithelial cells (HMECs). HMECs immortalized after chemical carcinogen exposure initially expressed little or no telomerase activity, and their telomeres continued to shorten with passage. Cell populations whose mean terminal restriction fragment (TRF) length declined to < or = 3 kb exhibited slow heterogeneous growth and contained many nonproliferative cells. These conditionally immortal HMEC cultures accumulated large quantities of p57 protein. With continued passage, the conditionally immortal cell populations very gradually converted to a fully immortal phenotype of good uniform growth, expression of high levels of telomerase activity, and stabilization of telomere length. The fully immortal HMECs that grew well did not accumulate p57 in G0 or during the cell cycle. DNA and RNA analysis of mass populations and individual subclones of conditionally immortal HMEC line 184A1 showed that continued growth of conditionally immortal cells with critically short telomeres was repeatedly accompanied by loss of the expressed p57 allele and transient expression of the allele imprinted previously. Conditionally immortal 184A1 with mean TRF > 3 kb, infected with retroviruses containing the p57 gene, exhibited premature slow heterogeneous growth. Conversely, exogenous expression of human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, in 184A1 with mean TRF > 3 kb prevented both the slow heterogeneous growth phase and accumulation of p57 in cycling populations. These data indicate that in HMECs that have overcome replicative senescence, p57 may provide an additional barrier against indefinite proliferation. Overcoming p57-mediated growth inhibition in these cells may be crucial for acquisition of the unlimited growth potential thought to be critical for malignant progression.
- Published
- 1999
41. Stability of tandem repeats in the Drosophila melanogaster Hsr-omega nuclear RNA.
- Author
-
Hogan NC, Slot F, Traverse KL, Garbe JC, Bendena WG, and Pardue ML
- Subjects
- Alleles, Animals, Base Sequence, Chromosome Mapping, Molecular Sequence Data, Oligodeoxyribonucleotides, Species Specificity, Cell Nucleus metabolism, Drosophila melanogaster genetics, RNA genetics, Repetitive Sequences, Nucleic Acid
- Abstract
The Drosophila melanogaster Hsr-omega locus produces a nuclear RNA containing > 5 kb of tandem repeat sequences. These repeats are unique to Hsr-omega and show concerted evolution similar to that seen with classical satellite DNAs. In D. melanogaster the monomer is approximately 280 bp. Sequences of 19 1/2 monomers differ by 8 +/- 5% (mean +/- SD), when all pairwise comparisons are considered. Differences are single nucleotide substitutions and 1-3 nucleotide deletions/insertions. Changes appear to be randomly distributed over the repeat unit. Outer repeats do not show the decrease in monomer homogeneity that might be expected if homogeneity is maintained by recombination. However, just outside the last complete repeat at each end, there are a few fragments of sequence similar to the monomer. The sequences in these flanking regions are not those predicted for sequences decaying in the absence of recombination. Instead, the fragmentation of the sequence homology suggests that flanking regions have undergone more severe disruptions, possibly during an insertion or amplification event. Hsr-omega alleles differing in the number of repeats are detected and appear to be stable over a few thousand generations; however, both increases and decreases in repeat numbers have been observed. The new alleles appear to be as stable as their predecessors. No alleles of less than approximately 5 kb nor more than approximately 16 kb of repeats were seen in any stocks examined. The evidence that there is a limit on the minimum number of repeats is consistent with the suggestion that these repeats are important in the function of the unusual Hsr-omega nuclear RNA.
- Published
- 1995
- Full Text
- View/download PDF
42. IMP-L2: an essential secreted immunoglobulin family member implicated in neural and ectodermal development in Drosophila.
- Author
-
Garbe JC, Yang E, and Fristrom JW
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Female, Molecular Sequence Data, Morphogenesis genetics, Phenotype, Sequence Alignment, Drosophila genetics, Ectoderm physiology, Genes, Insect genetics, Nervous System embryology
- Abstract
The Drosophila IMP-L2 gene was identified as a 20-hydroxyecdysone-induced gene encoding a membrane-bound polysomal transcript. IMP-L2 is an apparent secreted member of the immunoglobulin superfamily. We have used deficiencies that remove the IMP-L2 gene to demonstrate that IMP-L2 is essential in Drosophila. The viability of IMP-L2 null zygotes is influenced by maternal IMP-L2. IMP-L2 null progeny from IMP-L2+ mothers exhibit a semilethal phenotype. IMP-L2 null progeny from IMP-L2 null mothers are 100% lethal. An IMP-L2 transgene completely suppresses the zygotic lethal phenotype and partially suppresses the lethality of IMP-L2 null progeny from IMP-L2 null mothers. In embryos, IMP-L2 mRNA is first expressed at the cellular blastoderm stage and continues to be expressed through subsequent development. IMP-L2 mRNA is detected in several sites including the ventral neuroectoderm, the tracheal pits, the pharynx and esophagus, and specific neuronal cell bodies. Staining of whole-mount embryos with anti-IMP-L2 antibodies shows that IMP-L2 protein is localized to specific neuronal structures late in embryogenesis. Expression of IMP-L2 protein in neuronal cells suggests a role in the normal development of the nervous system but no severe morphological abnormalities have been detected in IMP-L2 null embryos.
- Published
- 1993
- Full Text
- View/download PDF
43. The Drosophila Stubble-stubbloid gene encodes an apparent transmembrane serine protease required for epithelial morphogenesis.
- Author
-
Appel LF, Prout M, Abu-Shumays R, Hammonds A, Garbe JC, Fristrom D, and Fristrom J
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Blotting, Northern, Cell Membrane enzymology, Chromosome Walking, Cloning, Molecular, DNA genetics, DNA isolation & purification, Drosophila melanogaster embryology, Drosophila melanogaster growth & development, Embryo, Nonmammalian, Epithelial Cells, Genes, Dominant, Genes, Recessive, Larva, Membrane Proteins metabolism, Molecular Sequence Data, Morphogenesis genetics, Protein Sorting Signals genetics, Pupa, RNA genetics, RNA isolation & purification, Sequence Homology, Amino Acid, Serine Endopeptidases metabolism, Transcription, Genetic, Drosophila Proteins, Drosophila melanogaster genetics, Membrane Proteins genetics, Serine Endopeptidases genetics
- Abstract
The Stubble-stubbloid (Sb-sbd) gene is required for hormone-dependent epithelial morphogenesis of imaginal discs of Drosophila, including the formation of bristles, legs, and wings. The gene has been cloned by using Sb-sbd-associated DNA lesions in a 20-kilobase (kb) region of a 263-kb genomic walk. The region specifies an approximately 3.8-kb transcript that is induced by the steroid hormone 20-hydroxyecdysone in imaginal discs cultured in vitro. The conceptually translated protein is an apparent 786-residue type II transmembrane protein (N terminus in, C terminus out), including an intracellular N-terminal domain of at least 35 residues and an extracellular C-terminal trypsin-like serine protease domain of 244 residues. Sequence analyses indicate that the Sb-sbd-encoded protease could activate itself by proteolytic cleavage. Consistent with the cell-autonomous nature of the Sb-sbd bristle phenotype, a disulfide bond between cysteine residues in the noncatalytic N-terminal fragment and the C-terminal catalytic fragment could tether the protease to the membrane after activation. Both dominant Sb and recessive sbd mutations affect the organization of microfilament bundles during bristle morphogenesis. We propose that the Sb-sbd product has a dual function. (i) It acts through its proteolytic extracellular domain to detach imaginal disc cells from extracellular matrices, and (ii) it transmits an outside-to-inside signal to its intracellular domain to modify the cytoskeleton and facilitate cell shape changes underlying morphogenesis.
- Published
- 1993
- Full Text
- View/download PDF
44. Expression of heat-shock locus hsr-omega in nonstressed cells during development in Drosophila melanogaster.
- Author
-
Bendena WG, Ayme-Southgate A, Garbe JC, and Pardue ML
- Subjects
- Animals, Cell Line, Cell Nucleus metabolism, Cytoplasm metabolism, Drosophila melanogaster genetics, Ecdysone pharmacology, Gene Expression Regulation drug effects, Homeostasis, Male, Nucleic Acid Hybridization, Spermatids physiology, Spermatocytes physiology, Spermatogenesis, Transcription, Genetic, Drosophila melanogaster embryology, Hot Temperature, RNA genetics
- Abstract
The hsr-omega locus forms one of the largest Drosophila heat-shock puffs and produces three major transcripts. These three transcripts are also produced constitutively, at lower levels, in almost all tissues and developmental stages. The amounts of the transcripts in nonstressed cells are modulated during development. The hormone ecdysone leads to increased levels of hsr-omega transcripts in cultured cells, suggesting that changing ecdysone titers may play a role in the developmental changes of hsr-omega transcript levels. By in situ hybridization to RNA in tissue sections, we detect only two cell types that lack hsr-omega transcripts--the preblastoderm embryo and the primary spermatocyte. There are no maternal transcripts of hsr-omega in the embryo. Transcripts appear abruptly at the time that the zygotic genome becomes transcriptionally active, shortly before the formation of the cellular blastoderm. No constitutive hsr-omega transcripts are found in primary spermatocytes. The spermatocytes cannot respond to heat shock by transcribing either hsr-omega or hsp70 RNA. Constitutive hsr-omega transcription is resumed later in spermatogenesis and hsr-omega RNA is detected in differentiating spermatids. These spermatids are also capable of mounting a heat-shock response, as measured by increases in hsr-omega and hsp70 RNA.
- Published
- 1991
- Full Text
- View/download PDF
45. Hsr-omega, A Novel Gene Encoded by a Drosophila Heat Shock Puff.
- Author
-
Pardue ML, Bendena WG, Fini ME, Garbe JC, Hogan NC, and Traverse KL
- Abstract
Although originally identified because of its abundant transcription in heat shock, the hsr-omega gene is active, at generally lower levels, in non-stressed cells. The locus produces an unusual set of three transcripts. Evidence from a variety of experiments suggests that one of these transcripts acts in the nucleus, possibly to regulate the activity of a nuclear protein. Another of the transcripts appears to act in the cytoplasm, possibly monitoring or regulating some aspect of translation. The two transcripts together could have a role in coordinating nuclear and cytoplasmic activity. A number of processes occur in eukaryotic cells in which nuclear and cytoplasmic activities need to be coordinated; we suggest that hsr-omega plays a role in such coordination.
- Published
- 1990
- Full Text
- View/download PDF
46. A Drosophila heat shock locus with a rapidly diverging sequence but a conserved structure.
- Author
-
Garbe JC, Bendena WG, Alfano M, and Pardue ML
- Subjects
- Animals, Base Sequence, Nucleic Acid Hybridization, Sequence Homology, Nucleic Acid, Drosophila genetics, Drosophila melanogaster genetics, Genes, Heat-Shock Proteins genetics
- Abstract
Cytological studies have shown that the heat shock loci 93D of Drosophila melanogaster and 2-48B of Drosophila hydei have several characteristics which suggest that they are homologous loci, yet sequence homology is barely detectable by cross-hybridization. Using cloned DNA sequences we have compared the two loci. Both loci produce transcripts of similar size and number. We have characterized the three predominant transcripts. In each species all three transcripts start at or about the same place within the unique portion of the gene. The longest transcript (approximately 9-10 kilobases) continues through several kilobases of short tandem repeats. The two shorter (1.9 and 1.2 kilobases) transcripts terminate 5' to the repeats. The repeat sequences are strongly conserved within a species but between species they have diverged both in length and in sequence. The longest homology is 9 nucleotides. The unique portions have also diverged significantly but do have some regions of conserved sequence. In both species the cytoplasmic transcript is spliced and polyadenylated but does not appear to contain a significant open reading frame. Thus, although the sequence of this locus has diverged significantly there are conserved features which suggest that the function of the locus is also conserved.
- Published
- 1986
47. MP13, a generalized transducing bacteriophage for Bacillus megaterium.
- Author
-
Vary PS, Garbe JC, Franzen M, and Frampton EW
- Subjects
- Bacteriophages physiology, Bacteriophages ultrastructure, Centrifugation, Density Gradient, Culture Media, DNA Restriction Enzymes, DNA, Viral, Temperature, Viral Plaque Assay, Bacillus megaterium genetics, Bacteriophages genetics, Transduction, Genetic
- Abstract
The first generalized transducing bacteriophage reported for Bacillus megaterium has been characterized. Optimum conditions for lysate production and transduction procedures were established so that transducing frequencies of 8 x 10(-6) and higher are now possible. The phage, MP13, has a head diameter of 97 nm and a contractile tail (202 by 17 nm) and adsorbs to the periphery of the cell. MP13 was inactivated rapidly at 60 degrees C, but not at 55 degrees C, and was sensitive to toluene, ether, and chloroform. When centrifuged in a neutral CsCl gradient, two bands were observed, a major band of 1.490 g cm-3 and a minor band of 1.482 g cm-3 buoyant density. The major band contained only infective particles, whereas the minor band contained both infective and transducing particles. Phage DNA was resistant to several restriction endonucleases, but yielded 9 fragments with MboI, more than 34 with HindIII, and 7 with BstEII. The molecular weights for the fragments from MboI-BstEII double digests total 97 x 10(9).
- Published
- 1982
- Full Text
- View/download PDF
48. Sequence evolution of the Drosophila heat shock locus hsr omega. I. The nonrepeated portion of the gene.
- Author
-
Garbe JC, Bendena WG, and Pardue ML
- Subjects
- Animals, Base Sequence, Drosophila genetics, Gene Expression Regulation, Hot Temperature, Molecular Sequence Data, Phylogeny, RNA biosynthesis, Species Specificity, Drosophila melanogaster genetics, Genes, Heat-Shock Proteins genetics, RNA genetics
- Abstract
The locus which we now call hsr omega was originally identified as a large heat shock puff in polytene region 93D of Drosophila melanogaster. This puff was subsequently found to have several phenotypic characteristics that distinguished it from other heat shock puffs. These characteristics include induction by a number of agents that do not induce other puffs and the presence of large ribonucleotide particles that are not found elsewhere. Each Drosophila species has one heat shock puff with these phenotypes. In contrast to the strong sequence conservation seen in puffs coding for heat shock proteins, very little cross-hybridization is detected between hsr omega loci in different species, suggesting that the hsr omega loci are diverging rapidly. Comparative analyses of the hsr omega locus from D. melanogaster, D. pseudoobscura, and D. hydei show that, despite the sequence change, the structure of the locus and its transcripts has been conserved, along with a number of short regions of the sequence. The short regions of conservation offer some clues to the function of this unusual locus. In addition, these comparisons offer a view of the evolution of a gene whose primary function does not appear to be protein coding.
- Published
- 1989
- Full Text
- View/download PDF
49. Heat shock: puffs and response to environmental stress.
- Author
-
Pardue ML, Bendena WG, and Garbe JC
- Subjects
- Animals, Base Sequence, Chromosomes ultrastructure, Gene Expression Regulation, Transcription, Genetic, Chromosomes physiology, Heat-Shock Proteins genetics, Hot Temperature
- Published
- 1987
- Full Text
- View/download PDF
50. Heat shock locus 93D of Drosophila melanogaster: a spliced RNA most strongly conserved in the intron sequence.
- Author
-
Garbe JC and Pardue ML
- Subjects
- Animals, Base Sequence, Cell Line, DNA genetics, Drosophila genetics, Genes, Genetic Markers, Peptides genetics, RNA, Messenger analysis, Sequence Homology, Nucleic Acid, Species Specificity, Drosophila melanogaster genetics, Heat-Shock Proteins genetics, RNA Splicing
- Abstract
The Drosophila melanogaster heat shock locus at 93D encodes at least three overlapping transcripts, 10-12 kilobases (kb), 1.9 kb, and 1.2 kb. The abundance of the three transcripts is significantly increased during heat shock; however, all are also found in non-heat-shocked cells. The 1.2-kb transcript is found in the cytoplasm. Sequence analysis of a 1.1-kb cDNA clone representing sequences within the 1.2-kb transcript and comparison to genomic sequences indicate that it is spliced; 700 base pairs of sequence found in genomic DNA are removed from the middle of the transcript. Sequence analysis further suggests that this RNA does not encode a heat shock protein. The largest open reading frame beginning with a methionine codon would encode a polypeptide of 34 amino acids. We have not been able to detect a heat shock-induced polypeptide of this size. A DNA clone from the analogous heat shock puff of Drosophila hydei has been analyzed by hybridization with the small subclones used to sequence the D. melanogaster cDNA plus a genomic fragment containing the 700-base-pair intron. Results of this hybridization indicated strong homology of the intron fragment. Weaker homology was detected with the two small fragments flanking the intron. Other fragments of the D. melanogaster cDNA showed no hybridization to the cloned D. hydei puff DNA.
- Published
- 1986
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.