Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Proc Natl Acad Sci U S A ; 121(11): e2307802121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437557

ABSTRACT

RNA interference (RNAi) therapeutics are an emerging class of medicines that selectively target mRNA transcripts to silence protein production and combat disease. Despite the recent progress, a generalizable approach for monitoring the efficacy of RNAi therapeutics without invasive biopsy remains a challenge. Here, we describe the development of a self-reporting, theranostic nanoparticle that delivers siRNA to silence a protein that drives cancer progression while also monitoring the functional activity of its downstream targets. Our therapeutic target is the transcription factor SMARCE1, which was previously identified as a key driver of invasion in early-stage breast cancer. Using a doxycycline-inducible shRNA knockdown in OVCAR8 ovarian cancer cells both in vitro and in vivo, we demonstrate that SMARCE1 is a master regulator of genes encoding proinvasive proteases in a model of human ovarian cancer. We additionally map the peptide cleavage profiles of SMARCE1-regulated proteases so as to design a readout for downstream enzymatic activity. To demonstrate the therapeutic and diagnostic potential of our approach, we engineered self-assembled layer-by-layer nanoparticles that can encapsulate nucleic acid cargo and be decorated with peptide substrates that release a urinary reporter upon exposure to SMARCE1-related proteases. In an orthotopic ovarian cancer xenograft model, theranostic nanoparticles were able to knockdown SMARCE1 which was in turn reported through a reduction in protease-activated urinary reporters. These LBL nanoparticles both silence gene products by delivering siRNA and noninvasively report on downstream target activity by delivering synthetic biomarkers to sites of disease, enabling dose-finding studies as well as longitudinal assessments of efficacy.


Subject(s)
Ovarian Neoplasms , Peptides , Humans , Female , RNA Interference , Peptides/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Peptide Hydrolases , RNA, Small Interfering/genetics , Endopeptidases , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins
2.
Diagnostics (Basel) ; 13(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36832208

ABSTRACT

The NavDx® blood test analyzes tumor tissue modified viral (TTMV)-HPV DNA to provide a reliable means of detecting and monitoring HPV-driven cancers. The test has been clinically validated in a large number of independent studies and has been integrated into clinical practice by over 1000 healthcare providers at over 400 medical sites in the US. This Clinical Laboratory Improvement Amendments (CLIA), high complexity laboratory developed test, has also been accredited by the College of American Pathologists (CAP) and the New York State Department of Health. Here, we report a detailed analytical validation of the NavDx assay, including sample stability, specificity as measured by limits of blank (LOBs), and sensitivity illustrated via limits of detection and quantitation (LODs and LOQs). LOBs were 0-0.32 copies/µL, LODs were 0-1.10 copies/µL, and LOQs were <1.20-4.11 copies/µL, demonstrating the high sensitivity and specificity of data provided by NavDx. In-depth evaluations including accuracy and intra- and inter-assay precision studies were shown to be well within acceptable ranges. Regression analysis revealed a high degree of correlation between expected and effective concentrations, demonstrating excellent linearity (R2 = 1) across a broad range of analyte concentrations. These results demonstrate that NavDx accurately and reproducibly detects circulating TTMV-HPV DNA, which has been shown to aid in the diagnosis and surveillance of HPV-driven cancers.

3.
Nat Commun ; 12(1): 7116, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893587

ABSTRACT

Mammary morphogenesis is an orchestrated process involving differentiation, proliferation and organization of cells to form a bi-layered epithelial network of ducts and lobules embedded in stromal tissue. We have engineered a 3D biomimetic human breast that makes it possible to study how stem cell fate decisions translate to tissue-level structure and function. Using this advancement, we describe the mechanism by which breast epithelial cells build a complex three-dimensional, multi-lineage tissue by signaling through a collagen receptor. Discoidin domain receptor tyrosine kinase 1 induces stem cells to differentiate into basal cells, which in turn stimulate luminal progenitor cells via Notch signaling to differentiate and form lobules. These findings demonstrate how human breast tissue regeneration is triggered by transmission of signals from the extracellular matrix through an epithelial bilayer to coordinate structural changes that lead to formation of a complex ductal-lobular network.


Subject(s)
Breast/cytology , Breast/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Discoidin Domain Receptor 1/metabolism , Biocompatible Materials , Biomedical Engineering , Cell Line , Discoidin Domain Receptor 1/genetics , Epithelial Cells/cytology , Extracellular Matrix , Humans , Regeneration , Signal Transduction , Stem Cells/cytology
4.
Cell Rep ; 28(2): 394-407.e6, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291576

ABSTRACT

DNA damage activates checkpoints that limit the replicative potential of stem cells, including differentiation. These checkpoints protect against cancer development but also promote tissue aging. Because mice lacking Slug/Snai2 exhibit limited stem cell activity, including luminobasal differentiation, and are protected from mammary cancer, we reasoned that Slug might regulate DNA damage checkpoints in mammary epithelial cells. Here, we show that Slug facilitates efficient execution of RPA32-mediated DNA damage response (DDR) signaling. Slug deficiency leads to delayed phosphorylation of ataxia telangiectasia mutated and Rad3-related protein (ATR) and its effectors RPA32 and CHK1. This leads to impaired RAD51 recruitment to DNA damage sites and persistence of unresolved DNA damage. In vivo, Slug/Snai2 loss leads to increased DNA damage and premature aging of mammary epithelium. Collectively, our work demonstrates that the mammary stem cell regulator Slug controls DDR checkpoints by dually inhibiting differentiation and facilitating DDR repair, and its loss causes unresolved DNA damage and accelerated aging.


Subject(s)
DNA Damage , DNA Repair , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Snail Family Transcription Factors/deficiency , Animals , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cellular Senescence/physiology , HEK293 Cells , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
5.
Cell Stem Cell ; 24(1): 65-78, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30554963

ABSTRACT

Our traditional understanding of phenotypic plasticity in adult somatic cells comprises dedifferentiation and transdifferentiation in the context of tissue regeneration or wound healing. Although dedifferentiation is central to tissue repair and stemness, this process inherently carries the risk of cancer initiation. Consequently, recent research suggests phenotypic plasticity as a new paradigm for understanding cancer initiation, progression, and resistance to therapy. Here, we discuss how cells acquire plasticity and the role of plasticity in initiating cancer, cancer progression, and metastasis and in developing therapy resistance. We also highlight the epithelial-to-mesenchymal transition (EMT) and known molecular mechanisms underlying plasticity and we consider potential therapeutic avenues.


Subject(s)
Adaptation, Physiological , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Disease Progression , Humans , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects
6.
Stem Cell Reports ; 10(3): 1131-1145, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29503088

ABSTRACT

The epithelial compartment of the mammary gland contains basal and luminal cell lineages, as well as stem and progenitor cells that reside upstream in the differentiation hierarchy. Stem and progenitor cell differentiation is regulated to maintain adult tissue and mediate expansion during pregnancy and lactation. The genetic factors that regulate the transition of cells between differentiation states remain incompletely understood. Here, we present a genome-scale method to discover genes driving cell-state specification. Applying this method, we identify a transcription factor, BCL11B, which drives stem cell self-renewal in vitro, by inhibiting differentiation into the basal lineage. To validate BCL11B's functional role, we use two-dimensional colony-forming and three-dimensional tissue differentiation assays to assess the lineage differentiation potential and functional abilities of primary human mammary cells. These findings show that BCL11B regulates mammary cell differentiation and demonstrate the utility of our proposed genome-scale strategy for identifying lineage regulators in mammalian tissues.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Self Renewal/genetics , Cell Self Renewal/physiology , Mammary Glands, Human/physiology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/physiology , Epithelial Cells/physiology , Female , Humans , Stem Cells/physiology , Transcription Factors/genetics
8.
Sci Rep ; 8(1): 1415, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362392

ABSTRACT

In cancer, tumor suppressor genes (TSGs) are frequently truncated, causing their encoded products to be non-functional or dominant-negative. We previously showed that premature polyadenylation (pPA) of MAGI3 truncates the gene, switching its functional role from a TSG to a dominant-negative oncogene. Here we report that MAGI3 undergoes pPA at the intron immediately downstream of its large internal exon, which is normally highly modified by N6-methyladenosine (m6A). In breast cancer cells that upregulate MAGI3 pPA , m6A levels in the large internal exon of MAGI3 are significantly reduced compared to cells that do not express MAGI3 pPA . We further find that MAGI3 pPA transcripts are significantly depleted of m6A modifications, in contrast to highly m6A-modified full-length MAGI3 mRNA. Finally, we analyze public expression data and find that other TSGs, including LATS1 and BRCA1, also undergo intronic pPA following large internal exons, and that m6A levels in these exons are reduced in pPA-activated breast cancer cells relative to untransformed mammary cells. Our study suggests that m6A may play a role in regulating intronic pPA of MAGI3 and possibly other TSGs, warranting further investigation.


Subject(s)
Adenosine/analogs & derivatives , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Adenosine/metabolism , BRCA1 Protein/metabolism , Cell Line, Tumor , Exons , Female , Gene Expression Regulation, Neoplastic , Humans , Introns , Membrane Proteins/metabolism , Polyadenylation , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Up-Regulation
9.
Nat Commun ; 8(1): 1079, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057869

ABSTRACT

PERK signaling is required for cancer invasion and there is interest in targeting this pathway for therapy. Unfortunately, chemical inhibitors of PERK's kinase activity cause on-target side effects that have precluded their further development. One strategy for resolving this difficulty would be to target downstream components of the pathway that specifically mediate PERK's pro-invasive and metastatic functions. Here we identify the transcription factor CREB3L1 as an essential mediator of PERK's pro-metastatic functions in breast cancer. CREB3L1 acts downstream of PERK, specifically in the mesenchymal subtype of triple-negative tumors, and its inhibition by genetic or pharmacological methods suppresses cancer cell invasion and metastasis. In patients with this tumor subtype, CREB3L1 expression is predictive of distant metastasis. These findings establish CREB3L1 as a key downstream mediator of PERK-driven metastasis and a druggable target for breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Breast Neoplasms/genetics , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Humans , Neoplasm Metastasis/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics
10.
Methods Mol Biol ; 1612: 139-147, 2017.
Article in English | MEDLINE | ID: mdl-28634940

ABSTRACT

We present a protocol for expanding human mammary tissues from primary patient-derived cells in three-dimensional (3D) cultures. The primary epithelial cells are seeded into 3D hydrogels with defined components, which include both proteins and carbohydrates present in mammary tissue. Over a span of 10-14 days, the seeded cells form mammary tissues with complex ductal-lobular topologies and include luminal and basal cells in the correct orientation, together with cells that stain positively for stem cell markers. In addition to recapitulating key architectural features of human mammary tissue, the expanded tissues also respond to lactogenic hormones including estrogen, progesterone, and prolactin. We anticipate that these cultures will prove useful for studies of mammary development and breast cancer.


Subject(s)
Breast/cytology , Cell Culture Techniques/methods , Epithelial Cells/cytology , Models, Biological , Cells, Cultured , Female , Humans , Hydrogels/chemistry , Tissue Engineering
11.
Proc Natl Acad Sci U S A ; 114(16): 4153-4158, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28377514

ABSTRACT

Advances in mammography have sparked an exponential increase in the detection of early-stage breast lesions, most commonly ductal carcinoma in situ (DCIS). More than 50% of DCIS lesions are benign and will remain indolent, never progressing to invasive cancers. However, the factors that promote DCIS invasion remain poorly understood. Here, we show that SMARCE1 is required for the invasive progression of DCIS and other early-stage tumors. We show that SMARCE1 drives invasion by regulating the expression of secreted proteases that degrade basement membrane, an ECM barrier surrounding all epithelial tissues. In functional studies, SMARCE1 promotes invasion of in situ cancers growing within primary human mammary tissues and is also required for metastasis in vivo. Mechanistically, SMARCE1 drives invasion by forming a SWI/SNF-independent complex with the transcription factor ILF3. In patients diagnosed with early-stage cancers, SMARCE1 expression is a strong predictor of eventual relapse and metastasis. Collectively, these findings establish SMARCE1 as a key driver of invasive progression in early-stage tumors.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Movement , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Recurrence, Local/pathology , Animals , Apoptosis , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cell Proliferation , Disease Progression , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Recurrence, Local/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Open Biol ; 7(2)2017 02.
Article in English | MEDLINE | ID: mdl-28202626

ABSTRACT

Phenotypic heterogeneity in cancers is associated with invasive progression and drug resistance. This heterogeneity arises in part from the ability of cancer cells to switch between phenotypic states, but the dynamics of this cellular plasticity remain poorly understood. Here we apply DNA barcodes to quantify and track phenotypic plasticity across hundreds of clones in a population of cancer cells exhibiting epithelial or mesenchymal differentiation phenotypes. We find that the epithelial-to-mesenchymal cell ratio is highly variable across the different clones in cancer cell populations, but remains stable for many generations within the progeny of any single clone-with a heritability of 0.89. To estimate the effects of combination therapies on phenotypically heterogeneous tumours, we generated quantitative simulations incorporating empirical data from our barcoding experiments. These analyses indicated that combination therapies which alternate between epithelial- and mesenchymal-specific treatments eventually select for clones with increased phenotypic plasticity. However, this selection could be minimized by increasing the frequency of alternation between treatments, identifying designs that may minimize selection for increased phenotypic plasticity. These findings establish new insights into phenotypic plasticity in cancer, and suggest design principles for optimizing the effectiveness of combination therapies for phenotypically heterogeneous tumours.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Neoplasm/genetics , Epithelial-Mesenchymal Transition , Neoplasms/genetics , Cell Line, Tumor , Cell Plasticity , DNA Barcoding, Taxonomic , Drug Resistance, Bacterial , Epithelial-Mesenchymal Transition/drug effects , HEK293 Cells , Humans , Neoplasm Invasiveness , Neoplasms/drug therapy , Neoplasms/pathology , Phenotype
13.
Proc Natl Acad Sci U S A ; 114(2): 382-387, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28028240

ABSTRACT

The use of proteasome inhibitors to target cancer's dependence on altered protein homeostasis has been greatly limited by intrinsic and acquired resistance. Analyzing data from thousands of cancer lines and tumors, we find that those with suppressed expression of one or more 19S proteasome subunits show intrinsic proteasome inhibitor resistance. Moreover, such proteasome subunit suppression is associated with poor outcome in myeloma patients, where proteasome inhibitors are a mainstay of treatment. Beyond conferring resistance to proteasome inhibitors, proteasome subunit suppression also serves as a sentinel of a more global remodeling of the transcriptome. This remodeling produces a distinct gene signature and new vulnerabilities to the proapoptotic drug, ABT-263. This frequent, naturally arising imbalance in 19S regulatory complex composition is achieved through a variety of mechanisms, including DNA methylation, and marks the emergence of a heritably altered and therapeutically relevant state in diverse cancers.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans
14.
Mol Cell ; 63(1): 60-71, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27320198

ABSTRACT

Despite its eponymous association with the heat shock response, yeast heat shock factor 1 (Hsf1) is essential even at low temperatures. Here we show that engineered nuclear export of Hsf1 results in cytotoxicity associated with massive protein aggregation. Genome-wide analysis revealed that Hsf1 nuclear export immediately decreased basal transcription and mRNA expression of 18 genes, which predominately encode chaperones. Strikingly, rescuing basal expression of Hsp70 and Hsp90 chaperones enabled robust cell growth in the complete absence of Hsf1. With the exception of chaperone gene induction, the vast majority of the heat shock response was Hsf1 independent. By comparative analysis of mammalian cell lines, we found that only heat shock-induced but not basal expression of chaperones is dependent on the mammalian Hsf1 homolog (HSF1). Our work reveals that yeast chaperone gene expression is an essential housekeeping mechanism and provides a roadmap for defining the function of HSF1 as a driver of oncogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , CRISPR-Cas Systems , Cell Line , DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Fungal , Gene Regulatory Networks , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Homeostasis , Mice, 129 Strain , Mice, Inbred CBA , Protein Aggregates , Protein Interaction Maps , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Time Factors , Transcription Factors/genetics , Transfection
15.
Breast Cancer Res ; 18(1): 19, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26926363

ABSTRACT

BACKGROUND: Three-dimensional (3D) cultures have proven invaluable for expanding human tissues for basic research and clinical applications. In both contexts, 3D cultures are most useful when they (1) support the outgrowth of tissues from primary human cells that have not been immortalized through extensive culture or viral infection and (2) include defined, physiologically relevant components. Here we describe a 3D culture system with both of these properties that stimulates the outgrowth of morphologically complex and hormone-responsive mammary tissues from primary human breast epithelial cells. METHODS: Primary human breast epithelial cells isolated from patient reduction mammoplasty tissues were seeded into 3D hydrogels. The hydrogel scaffolds were composed of extracellular proteins and carbohydrates present in human breast tissue and were cultured in serum-free medium containing only defined components. The physical properties of these hydrogels were determined using atomic force microscopy. Tissue growth was monitored over time using bright-field and fluorescence microscopy, and maturation was assessed using morphological metrics and by immunostaining for markers of stem cells and differentiated cell types. The hydrogel tissues were also studied by fabricating physical models from confocal images using a 3D printer. RESULTS: When seeded into these 3D hydrogels, primary human breast epithelial cells rapidly self-organized in the absence of stromal cells and within 2 weeks expanded to form mature mammary tissues. The mature tissues contained luminal, basal, and stem cells in the correct topological orientation and also exhibited the complex ductal and lobular morphologies observed in the human breast. The expanded tissues became hollow when treated with estrogen and progesterone, and with the further addition of prolactin produced lipid droplets, indicating that they were responding to hormones. Ductal branching was initiated by clusters of cells expressing putative mammary stem cell markers, which subsequently localized to the leading edges of the tissue outgrowths. Ductal elongation was preceded by leader cells that protruded from the tips of ducts and engaged with the extracellular matrix. CONCLUSIONS: These 3D hydrogel scaffolds support the growth of complex mammary tissues from primary patient-derived cells. We anticipate that this culture system will empower future studies of human mammary gland development and biology.


Subject(s)
Breast Neoplasms/pathology , Cell Culture Techniques , Cell Proliferation/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Female , Humans , Mammary Glands, Human/pathology , Stromal Cells/drug effects
16.
PLoS Comput Biol ; 11(4): e1004161, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25894653

ABSTRACT

The search for genes that regulate stem cell self-renewal and differentiation has been hindered by a paucity of markers that uniquely label stem cells and early progenitors. To circumvent this difficulty we have developed a method that identifies cell-state regulators without requiring any markers of differentiation, termed Perturbation-Expression Analysis of Cell States (PEACS). We have applied this marker-free approach to screen for transcription factors that regulate mammary stem cell differentiation in a 3D model of tissue morphogenesis and identified RUNX1 as a stem cell regulator. Inhibition of RUNX1 expanded bipotent stem cells and blocked their differentiation into ductal and lobular tissue rudiments. Reactivation of RUNX1 allowed exit from the bipotent state and subsequent differentiation and mammary morphogenesis. Collectively, our findings show that RUNX1 is required for mammary stem cells to exit a bipotent state, and provide a new method for discovering cell-state regulators when markers are not available.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Mammary Glands, Human/cytology , Stem Cells/cytology , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Profiling , Humans , Organoids/cytology , Organoids/metabolism , Systems Biology
17.
Elife ; 3: e03915, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25380226

ABSTRACT

The conserved Musashi (Msi) family of RNA binding proteins are expressed in stem/progenitor and cancer cells, but generally absent from differentiated cells, consistent with a role in cell state regulation. We found that Msi genes are rarely mutated but frequently overexpressed in human cancers and are associated with an epithelial-luminal cell state. Using ribosome profiling and RNA-seq analysis, we found that Msi proteins regulate translation of genes implicated in epithelial cell biology and epithelial-to-mesenchymal transition (EMT), and promote an epithelial splicing pattern. Overexpression of Msi proteins inhibited the translation of Jagged1, a factor required for EMT, and repressed EMT in cell culture and in mammary gland in vivo. Knockdown of Msis in epithelial cancer cells promoted loss of epithelial identity. Our results show that mammalian Msi proteins contribute to an epithelial gene expression program in neural and mammary cell types.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins/metabolism , Transcription, Genetic , Alternative Splicing/genetics , Animals , Base Sequence , Breast/growth & development , Breast/pathology , Breast Neoplasms/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Down-Regulation/genetics , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Ligands , Membrane Proteins/metabolism , Mice, Knockout , Models, Biological , Molecular Sequence Data , Morphogenesis , Neural Stem Cells/metabolism , Nucleotide Motifs/genetics , Protein Binding , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Receptors, Notch/metabolism , Serrate-Jagged Proteins
18.
PLoS Biol ; 12(9): e1001945, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25203443

ABSTRACT

Malignant carcinomas that recur following therapy are typically de-differentiated and multidrug resistant (MDR). De-differentiated cancer cells acquire MDR by up-regulating reactive oxygen species (ROS)-scavenging enzymes and drug efflux pumps, but how these genes are up-regulated in response to de-differentiation is not known. Here, we examine this question by using global transcriptional profiling to identify ROS-induced genes that are already up-regulated in de-differentiated cells, even in the absence of oxidative damage. Using this approach, we found that the Nrf2 transcription factor, which is the master regulator of cellular responses to oxidative stress, is preactivated in de-differentiated cells. In de-differentiated cells, Nrf2 is not activated by oxidation but rather through a noncanonical mechanism involving its phosphorylation by the ER membrane kinase PERK. In contrast, differentiated cells require oxidative damage to activate Nrf2. Constitutive PERK-Nrf2 signaling protects de-differentiated cells from chemotherapy by reducing ROS levels and increasing drug efflux. These findings are validated in therapy-resistant basal breast cancer cell lines and animal models, where inhibition of the PERK-Nrf2 signaling axis reversed the MDR of de-differentiated cancer cells. Additionally, analysis of patient tumor datasets showed that a PERK pathway signature correlates strongly with chemotherapy resistance, tumor grade, and overall survival. Collectively, these results indicate that de-differentiated cells up-regulate MDR genes via PERK-Nrf2 signaling and suggest that targeting this pathway could sensitize drug-resistant cells to chemotherapy.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , NF-E2-Related Factor 2/genetics , Neoplasm Recurrence, Local/genetics , eIF-2 Kinase/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cell Dedifferentiation/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred NOD , Mice, SCID , NF-E2-Related Factor 2/metabolism , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Oxidation-Reduction , Phosphorylation , Signal Transduction , Transcription, Genetic , eIF-2 Kinase/metabolism
19.
Stem Cell Reports ; 2(5): 633-47, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24936451

ABSTRACT

Perturbations in stem cell activity and differentiation can lead to developmental defects and cancer. We use an approach involving a quantitative model of cell-state transitions in vitro to gain insights into how SLUG/SNAI2, a key developmental transcription factor, modulates mammary epithelial stem cell activity and differentiation in vivo. In the absence of SLUG, stem cells fail to transition into basal progenitor cells, while existing basal progenitor cells undergo luminal differentiation; together, these changes result in abnormal mammary architecture and defects in tissue function. Furthermore, we show that in the absence of SLUG, mammary stem cell activity necessary for tissue regeneration and cancer initiation is lost. Mechanistically, SLUG regulates differentiation and cellular plasticity by recruiting the chromatin modifier lysine-specific demethylase 1 (LSD1) to promoters of lineage-specific genes to repress transcription. Together, these results demonstrate that SLUG plays a dual role in repressing luminal epithelial differentiation while unlocking stem cell transitions necessary for tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Disease-Free Survival , Gene Expression Regulation , Histones/metabolism , Humans , Mammary Glands, Human/cytology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Regeneration , Snail Family Transcription Factors , Stem Cell Transplantation , Stem Cells/cytology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transplantation, Heterologous
20.
Cancer Discov ; 4(6): 702-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24705811

ABSTRACT

UNLABELLED: Epithelial-to-mesenchymal transition (EMT) promotes both tumor progression and drug resistance, yet few vulnerabilities of this state have been identified. Using selective small molecules as cellular probes, we show that induction of EMT greatly sensitizes cells to agents that perturb endoplasmic reticulum (ER) function. This sensitivity to ER perturbations is caused by the synthesis and secretion of large quantities of extracellular matrix (ECM) proteins by EMT cells. Consistent with their increased secretory output, EMT cells display a branched ER morphology and constitutively activate the PERK-eIF2α axis of the unfolded protein response (UPR). Protein kinase RNA-like ER kinase (PERK) activation is also required for EMT cells to invade and metastasize. In human tumor tissues, EMT gene expression correlates strongly with both ECM and PERK-eIF2α genes, but not with other branches of the UPR. Taken together, our findings identify a novel vulnerability of EMT cells, and demonstrate that the PERK branch of the UPR is required for their malignancy. SIGNIFICANCE: EMT drives tumor metastasis and drug resistance, highlighting the need for therapies that target this malignant subpopulation. Our findings identify a previously unrecognized vulnerability of cancer cells that have undergone an EMT: sensitivity to ER stress. We also find that PERK-eIF2α signaling, which is required to maintain ER homeostasis, is also indispensable for EMT cells to invade and metastasize.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Epithelial-Mesenchymal Transition/genetics , Eukaryotic Initiation Factor-2/genetics , eIF-2 Kinase/genetics , Activating Transcription Factor 4/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice, SCID , Unfolded Protein Response , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...