Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nature ; 611(7935): 352-357, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36289331

ABSTRACT

The vertebrate adaptive immune system modifies the genome of individual B cells to encode antibodies that bind particular antigens1. In most mammals, antibodies are composed of heavy and light chains that are generated sequentially by recombination of V, D (for heavy chains), J and C gene segments. Each chain contains three complementarity-determining regions (CDR1-CDR3), which contribute to antigen specificity. Certain heavy and light chains are preferred for particular antigens2-22. Here we consider pairs of B cells that share the same heavy chain V gene and CDRH3 amino acid sequence and were isolated from different donors, also known as public clonotypes23,24. We show that for naive antibodies (those not yet adapted to antigens), the probability that they use the same light chain V gene is around 10%, whereas for memory (functional) antibodies, it is around 80%, even if only one cell per clonotype is used. This property of functional antibodies is a phenomenon that we call light chain coherence. We also observe this phenomenon when similar heavy chains recur within a donor. Thus, although naive antibodies seem to recur by chance, the recurrence of functional antibodies reveals surprising constraint and determinism in the processes of V(D)J recombination and immune selection. For most functional antibodies, the heavy chain determines the light chain.


Subject(s)
Antibodies , Clonal Selection, Antigen-Mediated , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Animals , Amino Acid Sequence , Antibodies/chemistry , Antibodies/genetics , Antibodies/immunology , Antigens/chemistry , Antigens/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mammals , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunologic Memory , V(D)J Recombination , Clonal Selection, Antigen-Mediated/genetics , Clonal Selection, Antigen-Mediated/immunology
2.
Anal Chem ; 90(2): 1273-1279, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29256243

ABSTRACT

Droplet microfluidics can identify and sort cells using digital reverse transcription polymerase chain reaction (RT-PCR) signals from individual cells. However, current methods require multiple microfabricated devices for enzymatic cell lysis and PCR reagent addition, making the process complex and prone to failure. Here, we describe a new approach that integrates all components into a single device. The method enables controlled exposure of isolated single cells to a high pH buffer, which lyses cells and inactivates reaction inhibitors but can be instantly neutralized with RT-PCR buffer. Using our chemical lysis approach, we distinguish individual cells' gene expression with data quality equivalent to more complex two-step workflows. Our system accepts cells and produces droplets ready for amplification, making single-cell droplet RT-PCR faster and more reliable.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Reverse Transcriptase Polymerase Chain Reaction/instrumentation , Single-Cell Analysis/instrumentation , Equipment Design , Gene Expression , Humans , Jurkat Cells , MCF-7 Cells
3.
Biomicrofluidics ; 11(4): 044107, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28794817

ABSTRACT

Droplet microfluidics can form and process millions of picoliter droplets with speed and ease, allowing the execution of huge numbers of biological reactions for high-throughput studies. However, at the conclusion of most experiments, the emulsions must be broken to recover and analyze their contents. This is usually achieved with demulsifiers, like perfluorooctanol and chloroform, which can interfere with downstream reactions and harm cells. Here, we describe a simple approach to rapidly and efficiently break microfluidic emulsions, which requires no chemicals. Our method allows one-pot multi-step reactions, making it useful for large scale automated processing of reactions requiring demulsification. Using a hand-held antistatic gun, we pulse emulsions with the electric field, coalescing ∼100 µl of droplets in ∼10 s. We show that while emulsions broken with chemical demulsifiers exhibit potent PCR inhibition, the antistatic-broken emulsions amplify efficiently. The ability to break emulsions quickly without chemicals should make our approach valuable for most demulsification needs in microfluidics.

4.
Proc Natl Acad Sci U S A ; 114(33): 8728-8733, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28760972

ABSTRACT

Although the elementary unit of biology is the cell, high-throughput methods for the microscale manipulation of cells and reagents are limited. The existing options either are slow, lack single-cell specificity, or use fluid volumes out of scale with those of cells. Here we present printed droplet microfluidics, a technology to dispense picoliter droplets and cells with deterministic control. The core technology is a fluorescence-activated droplet sorter coupled to a specialized substrate that together act as a picoliter droplet and single-cell printer, enabling high-throughput generation of intricate arrays of droplets, cells, and microparticles. Printed droplet microfluidics provides a programmable and robust technology to construct arrays of defined cell and reagent combinations and to integrate multiple measurement modalities together in a single assay.


Subject(s)
Microfluidic Analytical Techniques/methods , Microfluidics/methods , Biological Assay/methods , Cell Count/methods , Cell Line, Tumor , Humans , Printing/methods
5.
Proc Natl Acad Sci U S A ; 114(12): 3169-3174, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28258171

ABSTRACT

The transcription factor GATA3 is the master regulator that drives mammary luminal epithelial cell differentiation and maintains mammary gland homeostasis. Loss of GATA3 is associated with aggressive breast cancer development. We have identified ZNF503/ZEPPO2 zinc-finger elbow-related proline domain protein 2 (ZPO2) as a transcriptional repressor of GATA3 expression and transcriptional activity that induces mammary epithelial cell proliferation and breast cancer development. We show that ZPO2 is recruited to GATA3 promoter in association with ZBTB32 (Repressor of GATA, ROG) and that ZBTB32 is essential for down-regulation of GATA3 via ZPO2. Through this modulation of GATA3 activity, ZPO2 promotes aggressive breast cancer development. Our data provide insight into a mechanism of GATA3 regulation, and identify ZPO2 as a possible candidate gene for future diagnostic and therapeutic strategies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Animals , Binding Sites , Biopsy , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Cluster Analysis , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Mice , Neoplasm Metastasis , Promoter Regions, Genetic , Protein Binding
6.
Sci Rep ; 7: 44447, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28290550

ABSTRACT

Proteins are the primary effectors of cellular function, including cellular metabolism, structural dynamics, and information processing. However, quantitative characterization of proteins at the single-cell level is challenging due to the tiny amount of protein available. Here, we present Abseq, a method to detect and quantitate proteins in single cells at ultrahigh throughput. Like flow and mass cytometry, Abseq uses specific antibodies to detect epitopes of interest; however, unlike these methods, antibodies are labeled with sequence tags that can be read out with microfluidic barcoding and DNA sequencing. We demonstrate this novel approach by characterizing surface proteins of different cell types at the single-cell level and distinguishing between the cells by their protein expression profiles. DNA-tagged antibodies provide multiple advantages for profiling proteins in single cells, including the ability to amplify low-abundance tags to make them detectable with sequencing, to use molecular indices for quantitative results, and essentially limitless multiplexing.


Subject(s)
Dietary Proteins/analysis , Proteins/genetics , Single-Cell Analysis , DNA Barcoding, Taxonomic , Dietary Proteins/chemistry , High-Throughput Nucleotide Sequencing , Humans , Microfluidics/methods , Proteins/chemistry
7.
Mol Clin Oncol ; 6(1): 7-18, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123722

ABSTRACT

The achaete-scute complex-like (ASCL) family, also referred to as 'achaete-scute complex homolog' or 'achaete-scute family basic helix-loop-helix transcription factor', is critical for proper development of the nervous system and deregulation of ASCL plays a key role in psychiatric and neurological disorders. The ASCL family consists of five members, namely ASCL1, ASCL2, ASCL3, ASCL4 and ASCL5. The ASCL1 gene serves as a potential oncogene during lung cancer development. There is a correlation between increased ASCL2 expression and colon cancer development. Inhibition of ASCL2 reduced cellular proliferation and tumor growth in xenograft tumor experiments. Although previous studies demonstrated involvement of ASCL1 and ASCL2 in tumor development, little is known on the remaining ASCL family members and their potential effect on tumorigenesis. Therefore, a holistic approach to investigating the expression of ASCL family genes in diverse types of cancer may provide new insights in cancer research. In this study, we utilized a web-based microarray database (Oncomine; www.oncomine.org) to analyze the transcriptional expression of the ASCL family in clinical cancer and normal tissues. Our bioinformatics analysis revealed the potential involvement of multiple ASCL family members during tumor onset and progression in multiple types of cancer. Compared to normal tissue, ASCL1 exhibited a higher expression in cancers of the lung, pancreas, kidney, esophagus and head and neck, whereas ASCL2 exhibited a high expression in cancers of the breast, colon, stomach, lung, head and neck, ovary and testis. ASCL3, however, exhibited a high expression only in breast cancer. Interestingly, ASCL1 expression was downregulated in melanoma and in cancers of the bladder, breast, stomach and colon. ASCL2 exhibited low expression levels in sarcoma, melanoma, brain and prostate cancers. Reduction in the expression of ASCL3 was detected in lymphoma, bladder, cervical, kidney and epithelial cancers. Similarly, ASCL5 exhibited low expression in the majority of brain cancer subtypes, such as glioblastoma and oligodendroglioma. This analysis supports the hypothesis that specific ASCL members may play an important role in cancer development. Collectively, our data suggest that alterations in the expression of ASCL gene family members are correlated with cancer development. Furthermore, ASCL family members were categorized according to cancer subtype. The aim of this report was to provide novel insights to the significance of the ASCL family in various cancers and our findings suggested that the ASCL gene family may be an ideal target for future cancer studies.

8.
J Biol Chem ; 290(6): 3803-13, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25538248

ABSTRACT

The NET (nocA, Nlz, elB, TLP-1) subfamily of zinc finger proteins is an important mediator during developmental processes. The evolutionary conserved zinc finger protein ZNF503/Zeppo2 (zinc finger elbow-related proline domain protein 2, Zpo2) plays critical roles during embryogenesis. We found that Zpo2 is expressed in adult tissue and examined its function. We found that ZPO2 is a nuclearly targeted transcriptional repressor that is expressed in mammary epithelial cells. Elevated Zpo2 levels increase mammary epithelial cell proliferation. Zpo2 promotes cellular invasion through down-regulation of E-cadherin and regulates the invasive phenotype in a RAC1-dependent manner. We detect elevated Zpo2 expression during breast cancer progression in a MMTV-PyMT transgenic mouse model. Tumor transplant experiments indicated that overexpression of Zpo2 in MMTV-PyMT mammary tumor cell lines enhances lung metastasis. Our findings suggest that Zpo2 plays a significant role in mammary gland homeostasis and that deregulation of Zpo2 may promote breast cancer development.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Epithelial Cells/metabolism , Mammary Neoplasms, Experimental/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , Epithelial Cells/physiology , Female , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , rac1 GTP-Binding Protein/metabolism
9.
Cancer Res ; 74(2): 609-20, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24305876

ABSTRACT

The reactive stroma surrounding tumor lesions performs critical roles ranging from supporting tumor cell proliferation to inducing tumorigenesis and metastasis. Therefore, it is critical to understand the cellular components and signaling control mechanisms that underlie the etiology of reactive stroma. Previous studies have individually implicated fibroblast growth factor receptor 1 (FGFR1) and canonical WNT/ß-catenin signaling in prostate cancer progression and the initiation and maintenance of a reactive stroma; however, both pathways are frequently found to be coactivated in cancer tissue. Using autochthonous transgenic mouse models for inducible FGFR1 (JOCK1) and prostate-specific and ubiquitously expressed inducible ß-catenin (Pro-Cat and Ubi-Cat, respectively) and bigenic crosses between these lines (Pro-Cat × JOCK1 and Ubi-Cat × JOCK1), we describe WNT-induced synergistic acceleration of FGFR1-driven adenocarcinoma, associated with a pronounced fibroblastic reactive stroma activation surrounding prostatic intraepithelial neoplasia (mPIN) lesions found both in in situ and reconstitution assays. Both mouse and human reactive stroma exhibited increased transforming growth factor-ß (TGF-ß) signaling adjacent to pathologic lesions likely contributing to invasion. Furthermore, elevated stromal TGF-ß signaling was associated with higher Gleason scores in archived human biopsies, mirroring murine patterns. Our findings establish the importance of the FGFR1-WNT-TGF-ß signaling axes as driving forces behind reactive stroma in aggressive prostate adenocarcinomas, deepening their relevance as therapeutic targets.


Subject(s)
Prostatic Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Adenocarcinoma/metabolism , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Disease Models, Animal , Fibroblasts/metabolism , Humans , Inflammation , Male , Mice , Mice, Nude , Mice, Transgenic , Signal Transduction , Species Specificity , Stromal Cells/metabolism
10.
Cell Cycle ; 12(20): 3262-71, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24036551

ABSTRACT

The tumor microenvironment includes cells such as fibroblasts, immune cells, endothelial cells, as well as extracellular matrix (ECM), proteases, and cytokines. Together, these components participate in a complex crosstalk with neoplastic tumor cells that affects growth, angiogenesis, and metastasis. MicroRNAs (miRNAs) are small, non-coding RNAs involved in post-transcriptional regulation of gene expression and have recently emerged as important players involved in regulating multiple aspects of cancer biology and the tumor microenvironment. Differential miRNA expression in both the epithelial and stromal compartments of tumors compared with normal tissue suggests that miRNAs are important drivers of tumorigenesis and metastasis. This review article summarizes our current understanding of the diverse roles of miRNAs involved in tumor microenvironment regulation and underscores the importance of miRNAs within multiple cell types that contribute to the hallmarks of cancer.


Subject(s)
MicroRNAs/metabolism , Tumor Microenvironment/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology
11.
Cell Stem Cell ; 13(3): 300-13, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23871604

ABSTRACT

The microenvironment provides cues that control the behavior of epithelial stem and progenitor cells. Here, we identify matrix metalloproteinase-3 (MMP3) as a regulator of Wnt signaling and mammary stem cell (MaSC) activity. We show that MMP3 overexpression promotes hyperplastic epithelial growth, surprisingly, in a nonproteolytic manner via its hemopexin (HPX) domain. We demonstrate that MMP3-HPX specifically binds and inactivates Wnt5b, a noncanonical Wnt ligand that inhibits canonical Wnt signaling and mammary epithelial outgrowth in vivo. Indeed, transplants overexpressing MMP3 display increased canonical Wnt signaling, demonstrating that MMP3 is an extracellular regulator of the Wnt signaling pathway. MMP3-deficient mice exhibit decreased MaSC populations and diminished mammary-reconstituting activity, whereas MMP3 overexpression elevates MaSC function, indicating that MMP3 is necessary for the maintenance of MaSCs. Our study reveals a mechanism by a microenvironmental protease that regulates Wnt signaling and impacts adult epithelial stem cell function.


Subject(s)
Adult Stem Cells/physiology , Epithelium/physiology , Mammary Glands, Animal/cytology , Matrix Metalloproteinase 3/metabolism , Wnt Proteins/metabolism , Animals , Cells, Cultured , Cellular Microenvironment , Extracellular Matrix/metabolism , Hemopexin/metabolism , Matrix Metalloproteinase 3/genetics , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Protein Binding , Repressor Proteins/metabolism , Transgenes/genetics , Up-Regulation , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics
12.
Cell Stem Cell ; 11(5): 676-88, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23122291

ABSTRACT

The role of Notch signaling in the maintenance of adult murine prostate epithelial homeostasis remains unclear. We found that Notch ligands are mainly expressed within the basal cell lineage, while active Notch signaling is detected in both the prostate basal and luminal cell lineages. Disrupting the canonical Notch effector Rbp-j impairs the differentiation of prostate basal stem cells and increases their proliferation in vitro and in vivo, but does not affect luminal cell biology. Conversely, ectopic Notch activation in adult prostates results in a decrease in basal cell number and luminal cell hyperproliferation. TGFß dominates over Notch signaling and overrides Notch ablation-induced proliferation of prostate basal cells. However, Notch confers sensitivity and positive feedback by upregulating a plethora of TGFß signaling components including TgfßR1. These findings reveal crucial roles of the self-enforced positive reciprocal regulatory loop between TGFß and Notch in maintaining prostate basal stem cell dormancy.


Subject(s)
Prostate/cytology , Receptors, Notch/metabolism , Stem Cells/cytology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cell Proliferation , Male , Mice , Microscopy, Electron, Scanning Transmission , Prostate/metabolism , Signal Transduction , Stem Cells/metabolism
13.
PLoS One ; 7(1): e30814, 2012.
Article in English | MEDLINE | ID: mdl-22303459

ABSTRACT

Wnt signaling is crucial for a variety of biological processes, including body axis formation, planar polarity, stem cell maintenance and cellular differentiation. Therefore, targeted manipulation of Wnt signaling in vivo would be extremely useful. By applying chemical inducer of dimerization (CID) technology, we were able to modify the Wnt co-receptor, low-density lipoprotein (LDL)-receptor-related protein 5 (LRP5), to generate the synthetic ligand inducible Wnt switch, iLRP5. We show that iLRP5 oligomerization results in its localization to disheveled-containing punctate structures and sequestration of scaffold protein Axin, leading to robust ß-catenin-mediated signaling. Moreover, we identify a novel LRP5 cytoplasmic domain critical for its intracellular localization and casein kinase 1-dependent ß-catenin signaling. Finally, by utilizing iLRP5 as a Wnt signaling switch, we generated the Ubiquitous Activator of ß-catenin (Ubi-Cat) transgenic mouse line. The Ubi-Cat line allows for nearly ubiquitous expression of iLRP5 under control of the H-2K(b) promoter. Activation of iLRP5 in isolated prostate basal epithelial stem cells resulted in expansion of p63(+) cells and development of hyperplasia in reconstituted murine prostate grafts. Independently, iLRP5 induction in adult prostate stroma enhanced prostate tissue regeneration. Moreover, induction of iLRP5 in male Ubi-Cat mice resulted in prostate tumor progression over several months from prostate hyperplasia to adenocarcinoma. We also investigated iLRP5 activation in Ubi-Cat-derived mammary cells, observing that prolonged activation results in mammary tumor formation. Thus, in two distinct experimental mouse models, activation of iLRP5 results in disruption of tissue homeostasis, demonstrating the utility of iLRP5 as a novel research tool for determining the outcome of Wnt activation in a precise spatially and temporally determined fashion.


Subject(s)
Homeostasis , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Protein Multimerization , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Axin Protein/metabolism , Casein Kinase I/metabolism , Dishevelled Proteins , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Intracellular Space/metabolism , Male , Mammary Neoplasms, Animal/pathology , Membrane Microdomains/metabolism , Mice , Molecular Sequence Data , Phosphoproteins/metabolism , Phosphorylation , Prostate/pathology , Prostate/transplantation , Protein Binding , Protein Transport , Stromal Cells/metabolism , Stromal Cells/pathology , Structure-Activity Relationship , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
14.
EMBO Rep ; 12(9): 971-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21799517

ABSTRACT

The activation of innate and adaptive immunity is always balanced by inhibitory signalling mechanisms to maintain tissue integrity. We have identified the E3 ligase c-Cbl--known for its roles in regulating lymphocyte signalling--as a modulator of dendritic cell activation. In c-Cbl-deficient dendritic cells, Toll-like receptor-induced expression of proinflammatory factors, such as interleukin-12, is increased, correlating with a greater potency of dendritic-cell-based vaccines against established tumours. This proinflammatory phenotype is accompanied by an increase in nuclear factor (NF)-κB activity. In addition, c-Cbl deficiency reduces both p50 and p105 levels, which have been shown to modulate the stimulatory function of NF-κB. Our data indicate that c-Cbl has a crucial, RING-domain-dependent role in regulating dendritic cell maturation, probably by facilitating the regulatory function of p105 and/or p50.


Subject(s)
Dendritic Cells/immunology , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Animals , Antigens, Nuclear/biosynthesis , Antigens, Nuclear/metabolism , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/metabolism , Dendritic Cells/metabolism , Female , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Toll-Like Receptors/metabolism
15.
Stem Cells ; 29(4): 678-88, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21308863

ABSTRACT

Tissue stem cells are capable of both self-renewal and differentiation to maintain a constant stem cell population and give rise to the plurality of cells within a tissue. Wnt signaling has been previously identified as a key mediator for the maintenance of tissue stem cells; however, possible cross-regulation with other developmentally critical signaling pathways involved in adult tissue homeostasis, such as Notch, is not well understood. By using an in vitro prostate stem cell colony ("prostasphere") formation assay and in vivo prostate reconstitution experiments, we demonstrate that Wnt pathway induction on Sca-1(+) CD49f(+) basal/stem cells (B/SCs) promotes expansion of the basal epithelial compartment with noticeable increases in "triple positive" (cytokeratin [CK] 5(+), CK8(+), p63(+)) prostate progenitor cells, concomitant with upregulation of known Wnt target genes involved in cell-cycle induction. Moreover, Wnt induction affects expression of epithelial-to-mesenchymal transition signature genes, suggesting a possible mechanism for priming B/SC to act as potential tumor-initiating cells. Interestingly, induction of Wnt signaling in B/SCs results in downregulation of Notch1 transcripts, consistent with its postulated antiproliferative role in prostate cells. In contrast, induction of Notch signaling in prostate progenitors inhibits their proliferation and disrupts prostasphere formation. In vivo prostate reconstitution assays further demonstrate that induction of Notch in B/SCs disrupts proper acini formation in cells expressing the activated Notch1 allele, Notch-1 intracellular domain. These data emphasize the importance of Wnt/Notch cross-regulation in adult stem cell biology and suggest that Wnt signaling controls the proliferation and/or maintenance of epithelial progenitors via modulation of Notch signaling.


Subject(s)
Cell Differentiation , Cell Proliferation , Prostate/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Stem Cells/metabolism , Wnt Proteins/metabolism , Adult Stem Cells , Animals , Cell Cycle/genetics , Cell Line , Epithelial-Mesenchymal Transition , Fluorescent Antibody Technique , Male , Mice , Phosphoproteins , Prostate/cytology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Trans-Activators , Wnt Proteins/genetics , Wnt3 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...