551
|
Notch-1 signaling regulates astrocytic proliferation and activation after hypoxia exposure. Neurosci Lett 2015; 603:12-8. [DOI: 10.1016/j.neulet.2015.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 01/25/2023]
|
552
|
Notch signaling governs phenotypic modulation of smooth muscle cells. Vascul Pharmacol 2015; 63:88-96. [PMID: 25464923 DOI: 10.1016/j.vph.2014.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/22/2014] [Accepted: 09/16/2014] [Indexed: 01/27/2023]
Abstract
A feature of vascular smooth muscle cells is their unique ability to exist in multiple phenotypes permitting a broad range of functions that include contraction, proliferation, or synthesis and secretion of extracellular matrix. Although it is known that these phenotypes can be overlapping, the mechanisms that regulate phenotypic modulation are still unclear. Given that endothelial cells are known to convey signals to smooth muscle cells that govern their activities within the vasculature; we sought to better define how endothelial cells regulate phenotypic changes of smooth muscle cells in coculture conditions. Using human aortic smooth muscle cells, we show that endothelial cells promote an increase in a differentiated/contractile phenotype while decreasing proliferation. Analysis of the synthetic phenotype demonstrates that endothelial cells also increase collagen synthesis and secretion. Characterization of pathways important for these endothelial cell-dependent phenotypes reveal that Notch signaling plays an important role in the establishment of these smooth muscle properties. These data highlight the ability of endothelial cells to control phenotypic modulation in a unique and previously undefined manner.
Collapse
|
553
|
Cui D, Dai J, Keller JM, Mizokami A, Xia S, Keller ET. Notch Pathway Inhibition Using PF-03084014, a γ-Secretase Inhibitor (GSI), Enhances the Antitumor Effect of Docetaxel in Prostate Cancer. Clin Cancer Res 2015. [PMID: 26202948 DOI: 10.1158/1078-0432.ccr-15-0242] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the efficacy and mechanisms of Notch signaling inhibition as an adjuvant to docetaxel in castration-resistant prostate cancer (CRPC) using a γ-secretase inhibitor (GSI), PF-03084014. EXPERIMENTAL DESIGN The effect of PF-03084014 on response to docetaxel was evaluated in docetaxel-sensitive and docetaxel-resistant CRPC cell lines in vitro and in murine models. Both soft tissue and bone sites were evaluated in vivo. Impacts on cell proliferation, apoptosis, cancer stem cells, and angiogenesis were evaluated. RESULTS The combination of PF-03084014 plus docetaxel reduced both docetaxel-sensitive and docetaxel-resistant CRPC tumor growth in soft tissue and bone greater than either agent alone. Antitumor activity was associated with PF-03084014-induced inhibition of Notch pathway signaling; decreased survival signals (cyclin E; MEK/ERK, PI3K/AKT, EGFR and NF-κB pathway; BCL-2, BCL-XL); increased apoptotic signals (BAK, BAX; cleaved caspase-3); reduced microvessel density; reduced epithelial-mesenchymal transition; and reduced cancer stem-like cells in the tumor. CONCLUSIONS These results reveal that PF-03084014 enhances docetaxel-mediated tumor response and provides a rationale to explore GSIs as adjunct therapy in conjunction with docetaxel for men with CRPC.
Collapse
Affiliation(s)
- Di Cui
- Department of Urology, University of Michigan, Ann Arbor, Michigan. Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinlu Dai
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Jill M Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan. Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Shujie Xia
- Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan. Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
554
|
Hamada M, Goricki S, Byerly MS, Satoh N, Jeffery WR. Evolution of the chordate regeneration blastema: Differential gene expression and conserved role of notch signaling during siphon regeneration in the ascidian Ciona. Dev Biol 2015. [PMID: 26206613 DOI: 10.1016/j.ydbio.2015.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The regeneration of the oral siphon (OS) and other distal structures in the ascidian Ciona intestinalis occurs by epimorphosis involving the formation of a blastema of proliferating cells. Despite the longstanding use of Ciona as a model in molecular developmental biology, regeneration in this system has not been previously explored by molecular analysis. Here we have employed microarray analysis and quantitative real time RT-PCR to identify genes with differential expression profiles during OS regeneration. The majority of differentially expressed genes were downregulated during OS regeneration, suggesting roles in normal growth and homeostasis. However, a subset of differentially expressed genes was upregulated in the regenerating OS, suggesting functional roles during regeneration. Among the upregulated genes were key members of the Notch signaling pathway, including those encoding the delta and jagged ligands, two fringe modulators, and to a lesser extent the notch receptor. In situ hybridization showed a complementary pattern of delta1 and notch gene expression in the blastema of the regenerating OS. Chemical inhibition of the Notch signaling pathway reduced the levels of cell proliferation in the branchial sac, a stem cell niche that contributes progenitor cells to the regenerating OS, and in the OS regeneration blastema, where siphon muscle fibers eventually re-differentiate. Chemical inhibition also prevented the replacement of oral siphon pigment organs, sensory receptors rimming the entrance of the OS, and siphon muscle fibers, but had no effects on the formation of the wound epidermis. Since Notch signaling is involved in the maintenance of proliferative activity in both the Ciona and vertebrate regeneration blastema, the results suggest a conserved evolutionary role of this signaling pathway in chordate regeneration. The genes identified in this investigation provide the foundation for future molecular analysis of OS regeneration.
Collapse
Affiliation(s)
- Mayuko Hamada
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Spela Goricki
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Mardi S Byerly
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Noriyuki Satoh
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - William R Jeffery
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| |
Collapse
|
555
|
Orlova VV, Mummery CL. What Endothelial Cells from Patient iPSCs Can Tell Us about Aortic Valve Disease. Cell Stem Cell 2015; 16:455-7. [PMID: 25957898 DOI: 10.1016/j.stem.2015.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In a recent issue of Cell, Theodoris et al. (2015) used a complex systems biology approach to model vascular aortic calcification caused by mutations in the NOTCH1 gene. Comparison of endothelial cells from patient hiPSCs and genetically matched controls under fluid flow revealed novel mechanisms underlying the disease.
Collapse
Affiliation(s)
- Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
556
|
Jagged mediates differences in normal and tumor angiogenesis by affecting tip-stalk fate decision. Proc Natl Acad Sci U S A 2015; 112:E3836-44. [PMID: 26153421 DOI: 10.1073/pnas.1511814112] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is critical during development, wound repair, and cancer progression. During angiogenesis, some endothelial cells adopt a tip phenotype to lead the formation of new branching vessels; the trailing stalk cells proliferate to develop the vessel. Notch and VEGF signaling mediate the selection of these tip endothelial cells. However, how Jagged, a Notch ligand that is overexpressed in cancer, affects angiogenesis remains elusive. Here, by developing a theoretical framework for Notch-Delta-Jagged-VEGF signaling, we found that higher production levels of Jagged destabilizes the tip and stalk cell fates and can give rise to a hybrid tip/stalk phenotype that leads to poorly perfused and chaotic angiogenesis, which is a hallmark of cancer. Consistently, the signaling interactions that restrict Notch-Jagged signaling, such as Fringe, cis-inhibition, and increased production of Delta, stabilize tip and stalk fates and limit the existence of hybrid tip/stalk phenotype. Our results underline how overexpression of Jagged can transform physiological angiogenesis into pathological one.
Collapse
|
557
|
Li Y, Krahn JM, Flake GP, Umbach DM, Li L. Toward predicting metastatic progression of melanoma based on gene expression data. Pigment Cell Melanoma Res 2015; 28:453-63. [PMID: 25847062 PMCID: PMC4469521 DOI: 10.1111/pcmr.12374] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/27/2015] [Indexed: 01/20/2023]
Abstract
Primary and metastatic melanoma tumors share the same cell origin, making it challenging to identify genomic biomarkers that can differentiate them. Primary tumors themselves can be heterogeneous, reflecting ongoing genomic changes as they progress toward metastasizing. We developed a computational method to explore this heterogeneity and to predict metastatic progression of the primary tumors. We applied our method separately to gene expression and to microRNA (miRNA) expression data from ~450 primary and metastatic skin cutaneous melanoma (SKCM) samples from the Cancer Genome Atlas (TCGA). Metastatic progression scores from RNA-seq data were significantly associated with clinical staging of patients' lymph nodes, whereas scores from miRNA-seq data were significantly associated with Clark's level. The loss of expression of many characteristic epithelial lineage genes in primary SKCM tumor samples was highly correlated with predicted progression scores. We suggest that those genes/miRNAs might serve as putative biomarkers for SKCM metastatic progression.
Collapse
Affiliation(s)
- Yuanyuan Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Juno M. Krahn
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Gordon P. Flake
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - David M. Umbach
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Leping Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| |
Collapse
|
558
|
Barse L, Bocchetta M. Non-small-cell lung carcinoma: role of the Notch signaling pathway. LUNG CANCER (AUCKLAND, N.Z.) 2015; 6:43-53. [PMID: 28210150 PMCID: PMC5217522 DOI: 10.2147/lctt.s60329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Notch signaling plays a pivotal role during embryogenesis. It regulates three fundamental processes: lateral inhibition, boundary formation, and lineage specification. During post-natal life, Notch receptors and ligands control critical cell fate decisions both in compartments that are undergoing differentiation and in pluripotent progenitor cells. First recognized as a potent oncogene in certain lymphoblastic leukemias and mesothelium-derived tissue, the role of Notch signaling in epithelial, solid tumors has been far more controversial. The overall consequence of Notch signaling and which form of the Notch receptor drives malignancy in humans is deeply debated. Most likely, this is due to the high degree of context-dependent effects of Notch signaling. More recently, it has been discovered that Notch (especially Notch-1) can exert different, even opposite effects in the same tissue under differing microenvironmental conditions. Further complicating the understanding of Notch receptors is the recently discovered role for non-canonical Notch signaling. Additionally, the most frequent Notch signaling antagonists used in biological systems have been inhibitors of the transmembrane protease complex γ-secretase, which itself processes a plethora of class one transmembrane proteins and thus cannot be considered a Notch-specific upstream regulator. Here we review the available empirical evidence gathered in recent years concerning Notch receptors and ligands in non-small-cell lung carcinoma (NSCLC). Although an overview of the field reveals seemingly contradicting results, we propose that Notch signaling can be exploited as a therapeutic target in NSCLC and represents a promising complement to the current arsenal utilized to combat this malignancy, particularly in targeting NSCLC tissues under specific environmental conditions, such as hypoxia.
Collapse
Affiliation(s)
- Levi Barse
- Department of Pathology, Oncology Institute, Loyola University Chicago, Maywood, IL, USA
| | - Maurizio Bocchetta
- Department of Pathology, Oncology Institute, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
559
|
Chou MH, Hsieh YC, Huang CW, Chen PH, Chan SP, Tsao YP, Lee HH, Wu JT, Chen SL. BCAS2 Regulates Delta-Notch Signaling Activity through Delta Pre-mRNA Splicing in Drosophila Wing Development. PLoS One 2015; 10:e0130706. [PMID: 26091239 PMCID: PMC4475048 DOI: 10.1371/journal.pone.0130706] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 05/23/2015] [Indexed: 11/19/2022] Open
Abstract
Previously, we showed that BCAS2 is essential for Drosophila viability and functions in pre-mRNA splicing. In this study, we provide strong evidence that BCAS2 regulates the activity of Delta-Notch signaling via Delta pre-mRNA splicing. Depletion of dBCAS2 reduces Delta mRNA expression and leads to accumulation of Delta pre-mRNA, resulting in diminished transcriptions of Delta-Notch signaling target genes, such as cut and E(spl)m8. Furthermore, ectopic expression of human BCAS2 (hBCAS2) and Drosophila BCAS2 (dBCAS2) in a dBCAS2-deprived fly can rescue dBCAS2 depletion-induced wing damage to the normal phenotypes. These rescued phenotypes are correlated with the restoration of Delta pre-mRNA splicing, which affects Delta-Notch signaling activity. Additionally, overexpression of Delta can rescue the wing deformation by deprivation of dBCAS2; and the depletion of dBCAS2 can restore the aberrant eye associated with Delta-overexpressing retinas; providing supporting evidence for the regulation of Delta-Notch signaling by dBCAS2. Taken together, dBCAS2 participates in Delta pre-mRNA splicing that affects the regulation of Delta-Notch signaling in Drosophila wing development.
Collapse
Affiliation(s)
- Meng-Hsuan Chou
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yi-Chen Hsieh
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chu-Wei Huang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Po-Han Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Shih-Peng Chan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yeou-Ping Tsao
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, 104, Taiwan
| | - Hsiu-Hsiang Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - June-Tai Wu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, 100, Taiwan
- * E-mail: (SLC); (JTW)
| | - Show-Li Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- * E-mail: (SLC); (JTW)
| |
Collapse
|
560
|
Manning L, Weideman AM, Peercy B, Starz-Gaiano M. Tissue landscape alters adjacent cell fates during Drosophila egg development. Nat Commun 2015; 6:7356. [PMID: 26082073 PMCID: PMC4473798 DOI: 10.1038/ncomms8356] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 04/28/2015] [Indexed: 01/24/2023] Open
Abstract
Extracellular signalling molecules control many biological processes, but the influence of tissue architecture on the local concentrations of these factors is unclear. Here we examine this issue in the Drosophila egg chamber, where two anterior cells secrete Unpaired (Upd) to activate Signal transducer and activator of transcription (STAT) signalling in the epithelium. High STAT signalling promotes cell motility. Genetic analysis shows that all cells near the Upd source can respond. However, using upright imaging, we show surprising asymmetries in STAT activation patterns, suggesting that some cells experience different Upd levels than predicted by their location. We develop a three-dimensional mathematical model to characterize the spatio-temporal distribution of the activator. Simulations show that irregular tissue domains can produce asymmetric distributions of Upd, consistent with results in vivo. Mutant analysis substantiates this idea. We conclude that cellular landscape can heavily influence the effect of diffusible activators and should be more widely considered.
Collapse
Affiliation(s)
- Lathiena Manning
- Department of Biological Sciences, UMBC, 1000 Hilltop Circle, Baltimore, MD 21250
| | - Ann Marie Weideman
- Department of Mathematics and Statistics, UMBC, 1000 Hilltop Circle, Baltimore, MD 21250
| | - Bradford Peercy
- Department of Mathematics and Statistics, UMBC, 1000 Hilltop Circle, Baltimore, MD 21250
| | | |
Collapse
|
561
|
βA3/A1-crystallin and persistent fetal vasculature (PFV) disease of the eye. Biochim Biophys Acta Gen Subj 2015; 1860:287-98. [PMID: 26022148 DOI: 10.1016/j.bbagen.2015.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Persistent fetal vasculature (PFV) is a human disease in which the fetal vasculature of the eye fails to regress normally. The fetal, or hyaloid, vasculature nourishes the lens and retina during ocular development, subsequently regressing after formation of the retinal vessels. PFV causes serious congenital pathologies and is responsible for as much as 5% of blindness in the United States. SCOPE OF REVIEW The causes of PFV are poorly understood, however there are a number of animal models in which aspects of the disease are present. One such model results from mutation or elimination of the gene (Cryba1) encoding βA3/A1-crystallin. In this review we focus on the possible mechanisms whereby loss of functional βA3/A1-crystallin might lead to PFV. MAJOR CONCLUSIONS Cryba1 is abundantly expressed in the lens, but is also expressed in certain other ocular cells, including astrocytes. In animal models lacking βA3/A1-crystallin, astrocyte numbers are increased and they migrate abnormally from the retina to ensheath the persistent hyaloid artery. Evidence is presented that the absence of functional βA3/A1-crystallin causes failure of the normal acidification of endolysosomal compartments in the astrocytes, leading to impairment of certain critical signaling pathways, including mTOR and Notch/STAT3. GENERAL SIGNIFICANCE The findings suggest that impaired endolysosomal signaling in ocular astrocytes can cause PFV disease, by adversely affecting the vascular remodeling processes essential to ocular development, including regression of the fetal vasculature. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
|
562
|
Brockmann B, Mastel H, Oswald F, Maier D. Analysis of the interaction between human RITA and Drosophila Suppressor of Hairless. Hereditas 2015; 151:209-19. [PMID: 25588307 DOI: 10.1111/hrd2.00074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/01/2014] [Indexed: 10/24/2022] Open
Abstract
Notch signalling mediates intercellular communication, which is effected by the transcription factor CSL, an acronym for vertebrate CBF1/RBP-J, Drosophila Suppressor of Hairless [Su(H)] and C. elegans Lag1. Nuclear import of CBF1/RBP-J depends on co-activators and co-repressors, whereas the export relies on RITA. RITA is a tubulin and CBF1/RBP-J binding protein acting as a negative regulator of Notch signalling in vertebrates. RITA protein is highly conserved in eumatazoa, but no Drosophila homologue was yet identified. In this work, the activity of human RITA in the fly was addressed. To this end, we generated transgenic flies that allow a tissue specific induction of human RITA, which was demonstrated by Western blotting and in fly tissues. Unexpectedly, overexpression of RITA during fly development had little phenotypic consequences, even when overexpressed simultaneously with either Su(H) or the Notch antagonist Hairless. We demonstrate the in vivo binding of human RITA to Su(H) and to tubulin by co-immune precipitation. Moreover, RITA and tubulin co-localized to some degree in several Drosophila tissues. Overall our data show that human RITA, albeit binding to Drosophila Su(H) and tubulin, cannot influence the Notch signalling pathway in the fly, suggesting that a nuclear export mechanism of Su(H), if existent in Drosophila, does not depend on RITA.
Collapse
Affiliation(s)
- Birgit Brockmann
- Institute of Genetics, University of Hohenheim, Stuttgart, Germany
| | | | | | | |
Collapse
|
563
|
Bi P, Kuang S. Notch signaling as a novel regulator of metabolism. Trends Endocrinol Metab 2015; 26:248-55. [PMID: 25805408 PMCID: PMC4435535 DOI: 10.1016/j.tem.2015.02.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 12/11/2022]
Abstract
Evolutionarily unprepared for modern high-calorie diets and sedentary lifestyles, humans are now unprecedentedly susceptible to metabolic disorders such as obesity, type 2 diabetes (T2D), nonalcoholic fatty liver, and cardiovascular disease. These metabolic conditions are intertwined, together known as metabolic syndrome, and compromise human life quality as well as lives. Notch signaling, a fundamental signal transduction pathway critical for cell-cell communication and development, has recently been recognized as a key player in metabolism. This review summarizes the emerging roles of Notch signaling in regulating the metabolism of various cell and tissue types, with emphasis on the underlying molecular mechanisms and the potential of targeting this signal axis to treat metabolic diseases.
Collapse
Affiliation(s)
- Pengpeng Bi
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
564
|
Mutvei AP, Fredlund E, Lendahl U. Frequency and distribution of Notch mutations in tumor cell lines. BMC Cancer 2015; 15:311. [PMID: 25907971 PMCID: PMC4430925 DOI: 10.1186/s12885-015-1278-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/26/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Deregulated Notch signaling is linked to a variety of tumors and it is therefore important to learn more about the frequency and distribution of Notch mutations in a tumor context. METHODS In this report, we use data from the recently developed Cancer Cell Line Encyclopedia to assess the frequency and distribution of Notch mutations in a large panel of cancer cell lines in silico. RESULTS Our results show that the mutation frequency of Notch receptor and ligand genes is at par with that for established oncogenes and higher than for a set of house-keeping genes. Mutations were found across all four Notch receptor genes, but with notable differences between protein domains, mutations were for example more prevalent in the regions encoding the LNR and PEST domains in the Notch intracellular domain. Furthermore, an in silico estimation of functional impact showed that deleterious mutations cluster to the ligand-binding and the intracellular domains of NOTCH1. For most cell line groups, the mutation frequency of Notch genes is higher than in associated primary tumors. CONCLUSIONS Our results shed new light on the spectrum of Notch mutations after in vitro culturing of tumor cells. The higher mutation frequency in tumor cell lines indicates that Notch mutations are associated with a growth advantage in vitro, and thus may be considered to be driver mutations in a tumor cell line context.
Collapse
Affiliation(s)
| | - Erik Fredlund
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institute, SE-171 77, Stockholm, Sweden.
| | - Urban Lendahl
- Department of Cell and Molecular Biology, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
565
|
Bauer L, Takacs A, Slotta-Huspenina J, Langer R, Becker K, Novotny A, Ott K, Walch A, Hapfelmeier A, Keller G. Clinical Significance of NOTCH1 and NOTCH2 Expression in Gastric Carcinomas: An Immunohistochemical Study. Front Oncol 2015; 5:94. [PMID: 25954607 PMCID: PMC4406075 DOI: 10.3389/fonc.2015.00094] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 03/31/2015] [Indexed: 12/27/2022] Open
Abstract
Background NOTCH signaling can exert oncogenic or tumor suppressive functions and can contribute to chemotherapy resistance in cancer. In this study, we aimed to clarify the clinicopathological significance and the prognostic and predictive value of NOTCH1 and NOTCH2 expression in gastric cancer (GC). Methods NOTCH1 and NOTCH2 expression was determined immunohistochemically in 142 primarily resected GCs using tissue microarrays and in 84 pretherapeutic biopsies from patients treated by neoadjuvant chemotherapy. The results were correlated with survival, response to therapy, and clinico-pathological features. Results Primarily resected patients with NOTCH1-negative tumors demonstrated worse survival. High NOTCH1 expression was associated with early-stage tumors and with significantly increased survival in this subgroup. Higher NOTCH2 expression was associated with early-stage and intestinal-type tumors and with better survival in the subgroup of intestinal-type tumors. In pretherapeutic biopsies, higher NOTCH1 and NOTCH2 expression was more frequent in non-responding patients, but these differences were statistically not significant. Conclusion Our findings suggested that, in particular, NOTCH1 expression indicated good prognosis in GC. The close relationship of high NOTCH1 and NOTCH2 expression with early tumor stages may indicate a tumor-suppressive role of NOTCH signaling in GC. The role of NOTCH1 and NOTCH2 in neoadjuvantly treated GC is limited.
Collapse
Affiliation(s)
- Lukas Bauer
- Institute of Pathology, Technische Universität München , Munich , Germany
| | - Agnes Takacs
- Institute of Pathology, Technische Universität München , Munich , Germany
| | | | - Rupert Langer
- Department of Pathology, University of Bern , Bern , Switzerland
| | - Karen Becker
- Institute of Pathology, Technische Universität München , Munich , Germany
| | - Alexander Novotny
- Department of Surgery, Technische Universität München , Munich , Germany
| | - Katja Ott
- Department of Surgery, Klinikum Rosenheim , Rosenheim , Germany
| | - Axel Walch
- Institute of Pathology, Helmholtz-Zentrum München , Neuherberg , Germany
| | - Alexander Hapfelmeier
- Institute of Medical Statistics and Epidemiology, Technische Universität München , Munich , Germany
| | - Gisela Keller
- Institute of Pathology, Technische Universität München , Munich , Germany
| |
Collapse
|
566
|
Lillycrop KA, Costello PM, Teh AL, Murray RJ, Clarke-Harris R, Barton SJ, Garratt ES, Ngo S, Sheppard AM, Wong J, Dogra S, Burdge GC, Cooper C, Inskip HM, Gale CR, Gluckman PD, Harvey NC, Chong YS, Yap F, Meaney MJ, Rifkin-Graboi A, Holbrook JD, Godfrey KM. Association between perinatal methylation of the neuronal differentiation regulator HES1 and later childhood neurocognitive function and behaviour. Int J Epidemiol 2015; 44:1263-76. [PMID: 25906782 PMCID: PMC4588869 DOI: 10.1093/ije/dyv052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Early life environments induce long-term changes in neurocognitive development and behaviour. In animal models, early environmental cues affect neuropsychological phenotypes via epigenetic processes but, as yet, there is little direct evidence for such mechanisms in humans. Method We examined the relation between DNA methylation at birth and child neuropsychological outcomes in two culturally diverse populations using a genome-wide methylation analysis and validation by pyrosequencing. Results Within the UK Southampton Women’s Survey (SWS) we first identified 41 differentially methylated regions of interest (DMROI) at birth associated with child’s full-scale IQ at age 4 years. Associations between HES1 DMROI methylation and later cognitive function were confirmed by pyrosequencing in 175 SWS children. Consistent with these findings, higher HES1 methylation was associated with higher executive memory function in a second independent group of 200 SWS 7-year-olds. Finally, we examined a pathway for this relationship within a Singaporean cohort (n = 108). Here, HES1 DMROI methylation predicted differences in early infant behaviour, known to be associated with academic success. In vitro, methylation of HES1 inhibited ETS transcription factor binding, suggesting a functional role of this site. Conclusions Thus, our findings suggest that perinatal epigenetic processes mark later neurocognitive function and behaviour, providing support for a role of epigenetic processes in mediating the long-term consequences of early life environment on cognitive development.
Collapse
Affiliation(s)
- Karen A Lillycrop
- Centre for Biological Sciences, and NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK,
| | - Paula M Costello
- Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | - Robert J Murray
- Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
| | - Rebecca Clarke-Harris
- Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Emma S Garratt
- Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
| | - Sherry Ngo
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Allan M Sheppard
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Johnny Wong
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | - Shaillay Dogra
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | - Graham C Burdge
- Academic Unit of Human Development and Health, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK, NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK, NIHR Musculoskeletal Biomedical Research Unit, University of Oxford, Oxford, UK
| | - Hazel M Inskip
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Catharine R Gale
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Fabian Yap
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Duke NUS Graduate School of Medicine, National University of Singapore, Singapore, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore and
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, Canada
| | - Anne Rifkin-Graboi
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | - Joanna D Holbrook
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | | | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK, NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
567
|
Notch is a direct negative regulator of the DNA-damage response. Nat Struct Mol Biol 2015; 22:417-24. [PMID: 25895060 DOI: 10.1038/nsmb.3013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/23/2015] [Indexed: 11/08/2022]
Abstract
The DNA-damage response (DDR) ensures genome stability and proper inheritance of genetic information, both of which are essential to survival. It is presently unclear to what extent other signaling pathways modulate DDR function. Here we show that Notch receptor binds and inactivates ATM kinase and that this mechanism is evolutionarily conserved in Caenorhabditis elegans, Xenopus laevis and humans. In C. elegans, the Notch pathway impairs DDR signaling in gonad germ cells. In mammalian cells, activation of human Notch1 leads to reduced ATM signaling in a manner independent of Notch1 transcriptional activity. Notch1 binds directly to the regulatory FATC domain of ATM and inhibits ATM kinase activity. Notch1 and ATM activation are inversely correlated in human breast cancers, and inactivation of ATM by Notch1 contributes to the survival of Notch1-driven leukemia cells upon DNA damage.
Collapse
|
568
|
A novel transcriptional factor Nkapl is a germ cell-specific suppressor of Notch signaling and is indispensable for spermatogenesis. PLoS One 2015; 10:e0124293. [PMID: 25875095 PMCID: PMC4397068 DOI: 10.1371/journal.pone.0124293] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/11/2015] [Indexed: 02/06/2023] Open
Abstract
Spermatogenesis is an elaborately regulated system dedicated to the continuous production of spermatozoa via the genesis of spermatogonia. In this process, a variety of genes are expressed that are relevant to the differentiation of germ cells at each stage. Although Notch signaling plays a critical role in germ cell development in Drosophila and Caenorhabditis elegans, its function and importance for spermatogenesis in mammals is controversial. We report that Nkapl is a novel germ cell-specific transcriptional suppressor in Notch signaling. It is also associated with several molecules of the Notch corepressor complex such as CIR, HDAC3, and CSL. It was expressed robustly in spermatogonia and early spermatocytes after the age of 3 weeks. Nkapl-deleted mice showed complete arrest at the level of pachytene spermatocytes. In addition, apoptosis was observed in this cell type. Overexpression of NKAPL in germline stem cells demonstrated that Nkapl induced changes in spermatogonial stem cell (SSC) markers and the reduction of differentiation factors through the Notch signaling pathway, whereas testes with Nkapl deleted showed inverse changes in those markers and factors. Therefore, Nkapl is indispensable because aberrantly elevated Notch signaling has negative effects on spermatogenesis, affecting SSC maintenance and differentiation factors. Notch signaling should be properly regulated through the transcriptional factor Nkapl.
Collapse
|
569
|
Wang H, Zang C, Liu XS, Aster JC. The role of Notch receptors in transcriptional regulation. J Cell Physiol 2015; 230:982-8. [PMID: 25418913 DOI: 10.1002/jcp.24872] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/19/2014] [Indexed: 12/22/2022]
Abstract
Notch signaling has pleiotropic context-specific functions that have essential roles in many processes, including embryonic development and maintenance and homeostasis of adult tissues. Aberrant Notch signaling (both hyper- and hypoactive) is implicated in a number of human developmental disorders and many cancers. Notch receptor signaling is mediated by tightly regulated proteolytic cleavages that lead to the assembly of a nuclear Notch transcription complex, which drives the expression of downstream target genes and thereby executes Notch's functions. Thus, understanding regulation of gene expression by Notch is central to deciphering how Notch carries out its many activities. Here, we summarize the recent findings pertaining to the complex interplay between the Notch transcriptional complex and interacting factors involved in transcriptional regulation, including co-activators, cooperating transcription factors, and chromatin regulators, and discuss emerging data pertaining to the role of Notch-regulated noncoding RNAs in transcription.
Collapse
Affiliation(s)
- Hongfang Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | |
Collapse
|
570
|
Schwanbeck R. The role of epigenetic mechanisms in Notch signaling during development. J Cell Physiol 2015; 230:969-81. [PMID: 25336183 DOI: 10.1002/jcp.24851] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 10/16/2014] [Indexed: 12/16/2022]
Abstract
The Notch pathway is a highly conserved cell-cell communication pathway in metazoan involved in numerous processes during embryogenesis, development, and adult organisms. Ligand-receptor interaction of Notch components on adjacent cells facilitates controlled sequential proteolytic cleavage resulting in the nuclear translocation of the intracellular domain of Notch (NICD). There it binds to the Notch effector protein RBP-J, displaces a corepressor complex and enables the induction of target genes by recruitment of coactivators in a cell-context dependent manner. Both, the gene-specific repression and the context dependent activation require an intense communication with the underlying chromatin of the regulatory regions. Since the epigenetic landscape determines the function of the genome, processes like cell fate decision, differentiation, and self-renewal depend on chromatin structure and its remodeling during development. In this review, structural features enabling the Notch pathway to read these epigenetic marks by proteins interacting with RBP-J/Notch will be discussed. Furthermore, mechanisms of the Notch pathway to write and erase chromatin marks like histone acetylation and methylation are depicted as well as ATP-dependent chromatin remodeling during the activation of target genes. An additional fine-tuning of transcriptional regulation upon Notch activation seems to be controlled by the commitment of miRNAs. Since cells within an organism have to react to environmental changes, and developmental and differentiation cues in a proper manner, different signaling pathways have to crosstalk to each other. The chromatin status may represent one major platform to integrate these different pathways including the canonical Notch signaling.
Collapse
Affiliation(s)
- Ralf Schwanbeck
- Institute of Biochemistry, Medical Faculty, University of Kiel, Kiel, Germany
| |
Collapse
|
571
|
Kiriakidis S, Henze A, Kruszynska‐Ziaja I, Skobridis K, Theodorou V, Paleolog EM, Mazzone M. Factor‐inhibiting HIF‐1 (FIH‐1) is required for human vascular endothelial cell survival. FASEB J 2015; 29:2814-27. [DOI: 10.1096/fj.14-252379] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 03/06/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Serafim Kiriakidis
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesKennedy Institute of Rheumatology, University of OxfordUnited Kingdom
| | - Anne‐Theres Henze
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB)LeuvenBelgium
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research CenterDepartment of OncologyKatholieke Universiteit (KU)LeuvenBelgium
| | - Ilona Kruszynska‐Ziaja
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesKennedy Institute of Rheumatology, University of OxfordUnited Kingdom
| | - Konstantinos Skobridis
- Department of ChemistrySection of Organic Chemistry and BiochemistryUniversity of IoanninaGreece
| | - Vassiliki Theodorou
- Department of ChemistrySection of Organic Chemistry and BiochemistryUniversity of IoanninaGreece
| | - Ewa M. Paleolog
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesKennedy Institute of Rheumatology, University of OxfordUnited Kingdom
| | - Massimiliano Mazzone
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB)LeuvenBelgium
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research CenterDepartment of OncologyKatholieke Universiteit (KU)LeuvenBelgium
| |
Collapse
|
572
|
Chip physically interacts with Notch and their stoichiometry is critical for Notch function in wing development and cell proliferation in Drosophila. Biochim Biophys Acta Gen Subj 2015; 1850:802-12. [DOI: 10.1016/j.bbagen.2014.12.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/15/2014] [Accepted: 12/27/2014] [Indexed: 12/17/2022]
|
573
|
Placing ion channels into a signaling network of T cells: from maturing thymocytes to healthy T lymphocytes or leukemic T lymphoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:750203. [PMID: 25866806 PMCID: PMC4383400 DOI: 10.1155/2015/750203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
T leukemogenesis is a multistep process, where the genetic errors during T cell maturation cause the healthy progenitor to convert into the leukemic precursor that lost its ability to differentiate but possesses high potential for proliferation, self-renewal, and migration. A new misdirecting "leukemogenic" signaling network appears, composed by three types of participants which are encoded by (1) genes implicated in determined stages of T cell development but deregulated by translocations or mutations, (2) genes which normally do not participate in T cell development but are upregulated, and (3) nondifferentially expressed genes which become highly interconnected with genes expressed differentially. It appears that each of three groups may contain genes coding ion channels. In T cells, ion channels are implicated in regulation of cell cycle progression, differentiation, activation, migration, and cell death. In the present review we are going to reveal a relationship between different genetic defects, which drive the T cell neoplasias, with calcium signaling and ion channels. We suggest that changes in regulation of various ion channels in different types of the T leukemias may provide the intracellular ion microenvironment favorable to maintain self-renewal capacity, arrest differentiation, induce proliferation, and enhance motility.
Collapse
|
574
|
|
575
|
Combining detection of Notch1 and tumor necrosis factor-α converting enzyme is a reliable biomarker for the diagnosis of abdominal aortic aneurysms. Life Sci 2015; 127:39-45. [PMID: 25744398 DOI: 10.1016/j.lfs.2015.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/12/2015] [Accepted: 02/06/2015] [Indexed: 11/20/2022]
Abstract
AIMS Although many markers were associated with abdominal aortic aneurysm (AAA), there is no clear consensus on which marker is of the most value. Studies have implicated the role of Notch signaling in the pathogenesis of AAA. We investigate the value of plasma Jagged1, Notch receptors and tumor necrosis factor-α converting enzyme (TACE) in identifying AAA. MAIN METHODS 42 patients with AAA and 36 controls were enrolled in our study. The concentrations of plasma Jagged1, Notch receptors and TACE were measured by enzyme-linked immunosorbent assay (ELISA). The diagnostic value of plasma Notch1 and TACE was assessed by logistic regression and receiver operator characteristic (ROC) curve. Double immunofluorescence staining was used to investigate the distribution of Notch1 and TACE in AAA tissue specimens. KEY FINDINGS The concentrations of plasma Notch1 and TACE were significantly higher in AAA than in the controls, respectively (Notch1: P < 0.001; TACE: P = 0.0001). The area under the curve (AUC) from ROC curve of plasma Notch1 and TACE in determining the presence of AAA was 0.878 and 0.804, respectively. Combining detection of plasma Notch1 and TACE could improve the accuracy in detecting AAA (AUC 0.984, P < 0.0001). The predicted probability cutoff of 0.70 gave a sensitivity of 90.5% and a specificity of 100% for combining detection of plasma Notch1 and TACE in predicting AAA. SIGNIFICANCE This is the first report revealing that plasma Notch1 and TACE are highly expressed in AAA. Combining detection of plasma Notch1 and TACE may be reliable for identifying the presence of AAA.
Collapse
|
576
|
Beazley KE, Nurminsky D, Lima F, Gandhi C, Nurminskaya MV. Wnt16 attenuates TGFβ-induced chondrogenic transformation in vascular smooth muscle. Arterioscler Thromb Vasc Biol 2015; 35:573-9. [PMID: 25614285 PMCID: PMC4344425 DOI: 10.1161/atvbaha.114.304393] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Phenotypic plasticity of vascular smooth muscle cells (VSMCs) contributes to cardiovascular disease. Chondrocyte-like transformation of VSMCs associates with vascular calcification and underlies the formation of aortic cartilaginous metaplasia induced in mice by genetic loss of matrix Gla protein (MGP). Previous microarray analysis identified a dramatic downregulation of Wnt16 in calcified MGP-null aortae, suggesting an antagonistic role for Wnt16 in the chondrogenic transformation of VSMCs. APPROACH AND RESULTS Wnt16 is significantly downregulated in MGP-null aortae, before the histological appearance of cartilaginous metaplasia, and in primary MGP-null VSMCs. In contrast, intrinsic TGFβ is activated in MGP-null VSMCs and is necessary for spontaneous chondrogenesis of these cells in high-density micromass cultures. TGFβ3-induced chondrogenic transformation in wild-type VSMCs associates with Smad2/3-dependent Wnt16 downregulation, but Wnt16 does not suppress TGFβ3-induced Smad activation. In addition, TGFβ3 inhibits Notch signaling in wild-type VSMCs, and this pathway is downregulated in MGP-null aortae. Exogenous Wnt16 stimulates Notch activity and attenuates TGFβ3-induced downregulation of Notch in wild-type VSMCs, prevents chondrogenesis in MGP-null and TGFβ3-treated wild-type VSMCs, and stabilizes expression of contractile markers of differentiated VSMCs. CONCLUSIONS We describe a novel TGFβ-Wnt16-Notch signaling conduit in the chondrocyte-like transformation of VSMCs and identify endogenous TGFβ activity in MGP-null VSMCs as a critical mediator of chondrogenesis. Our proposed model suggests that the activated TGFβ pathway inhibits expression of Wnt16, which is a positive regulator of Notch signaling and a stabilizer of VSMC phenotype. These data advance the comprehensive mechanistic understanding of VSMC transformation and may identify a novel potential therapeutic target in vascular calcification.
Collapse
Affiliation(s)
- Kelly E Beazley
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Dmitry Nurminsky
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Florence Lima
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Chintan Gandhi
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Maria V Nurminskaya
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore.
| |
Collapse
|
577
|
Suresh S, Irvine AE. The NOTCH signaling pathway in normal and malignant blood cell production. J Cell Commun Signal 2015; 9:5-13. [PMID: 25711903 PMCID: PMC4414835 DOI: 10.1007/s12079-015-0271-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/10/2015] [Indexed: 12/23/2022] Open
Abstract
The NOTCH pathway is an evolutionarily conserved signalling network, which is fundamental in regulating developmental processes in invertebrates and vertebrates (Gazave et al. in BMC Evol Biol 9:249, 2009). It regulates self-renewal (Butler et al. in Cell Stem Cell 6:251-264, 2010), differentiation (Auderset et al. in Curr Top Microbiol Immunol 360:115-134, 2012), proliferation (VanDussen et al. in Development 139:488-497, 2012) and apoptosis (Cao et al. in APMIS 120:441-450, 2012) of diverse cell types at various stages of their development. NOTCH signalling governs cell-cell interactions and the outcome of such responses is highly context specific. This makes it impossible to generalize about NOTCH functions as it stimulates survival and differentiation of certain cell types, whereas inhibiting these processes in others (Meier-Stiegen et al. in PLoS One 5:e11481, 2010). NOTCH was first identified in 1914 in Drosophila and was named after the indentations (notches) present in the wings of the mutant flies (Bigas et al. in Int J Dev Biol 54:1175-1188, 2010). Homologs of NOTCH in vertebrates were initially identified in Xenopus (Coffman et al. in Science 249:1438-1441, 1990) and in humans NOTCH was first identified in T-Acute Lymphoblastic Leukaemia (T-ALL) (Ellisen et al. in Cell 66:649-61, 1991). NOTCH signalling is integral in neurogenesis (Mead and Yutzey in Dev Dyn 241:376-389, 2012), myogenesis (Schuster-Gossler et al. in Proc Natl Acad Sci U S A 104:537-542, 2007), haematopoiesis (Bigas et al. in Int J Dev Biol 54:1175-1188, 2010), oogenesis (Xu and Gridley in Genet Res Int 2012:648207, 2012), differentiation of intestinal cells (Okamoto et al. in Am J Physiol Gastrointest Liver Physiol 296:G23-35, 2009) and pancreatic cells (Apelqvist et al. in Nature 400:877-881, 1999). The current review will focus on NOTCH signalling in normal and malignant blood cell production or haematopoiesis.
Collapse
Affiliation(s)
- Sukanya Suresh
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | |
Collapse
|
578
|
Expansion of stem cells counteracts age-related mammary regression in compound Timp1/Timp3 null mice. Nat Cell Biol 2015; 17:217-27. [PMID: 25706237 DOI: 10.1038/ncb3118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 01/19/2015] [Indexed: 12/17/2022]
Abstract
Age is the primary risk factor for breast cancer in women. Bipotent basal stem cells actively maintain the adult mammary ductal tree, but with age tissues atrophy. We show that cell-extrinsic factors maintain the adult stem cell pool during ageing and dictate tissue stoichiometry. Mammary stem cells spontaneously expand more than 11-fold in virgin adult female mice lacking specific genes for TIMPs, the natural metalloproteinase inhibitors. Compound Timp1/Timp3 null glands exhibit Notch activation and accelerated gestational differentiation. Proteomics of mutant basal cells uncover altered cytoskeletal and extracellular protein repertoires, and we identify aberrant mitotic spindle orientation in these glands, a process that instructs asymmetric cell division and fate. We find that progenitor activity normally declines with age, but enriched stem/progenitor pools prevent tissue regression in Timp mutant mammary glands without affecting carcinogen-induced cancer susceptibility. Thus, improved stem cell content can extend mouse mammary tissue lifespan without altering cancer risk in this mouse model.
Collapse
|
579
|
Abstract
The latest discoveries and advanced knowledge in the fields of stem cell biology and developmental cardiology hold great promise for cardiac regenerative medicine, enabling researchers to design novel therapeutic tools and approaches to regenerate cardiac muscle for diseased hearts. However, progress in this arena has been hampered by a lack of reproducible and convincing evidence, which at best has yielded modest outcomes and is still far from clinical practice. To address current controversies and move cardiac regenerative therapeutics forward, it is crucial to gain a deeper understanding of the key cellular and molecular programs involved in human cardiogenesis and cardiac regeneration. In this review, we consider the fundamental principles that govern the "programming" and "reprogramming" of a human heart cell and discuss updated therapeutic strategies to regenerate a damaged heart.
Collapse
Affiliation(s)
- Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden Department of Medicine-Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Federica Santoro
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden Department of Medicine-Cardiology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
580
|
Activation of NOTCH1 or NOTCH3 signaling skews human airway basal cell differentiation toward a secretory pathway. PLoS One 2015; 10:e0116507. [PMID: 25700162 PMCID: PMC4336283 DOI: 10.1371/journal.pone.0116507] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 12/10/2014] [Indexed: 12/22/2022] Open
Abstract
Airway basal cells (BC) function as stem/progenitor cells capable of differentiating into the luminal ciliated and secretory cells to replenish the airway epithelium during physiological turnover and repair. The objective of this study was to define the role of Notch signaling in regulating human airway BC differentiation into a pseudostratified mucociliated epithelium. Notch inhibition with γ-secretase inhibitors demonstrated Notch activation is essential for BC differentiation into secretory and ciliated cells, but more so for the secretory lineage. Sustained cell autonomous ligand independent Notch activation via lentivirus expression of the intracellular domain of each Notch receptor (NICD1-4) demonstrated that the NOTCH2 and 4 pathways have little effect on BC differentiation into secretory and ciliated cells, while activation of the NOTCH1 or 3 pathways has a major influence, with persistent expression of NICD1 or 3 resulting in a skewing toward secretory cell differentiation with a parallel decrease in ciliated cell differentiation. These observations provide insights into the control of the balance of BC differentiation into the secretory vs ciliated cell lineage, a balance that is critical for maintaining the normal function of the airway epithelium in barrier defense against the inhaled environment.
Collapse
|
581
|
Abetov D, Mustapova Z, Saliev T, Bulanin D. Biomarkers and signaling pathways of colorectal cancer stem cells. Tumour Biol 2015; 36:1339-53. [PMID: 25680406 DOI: 10.1007/s13277-015-3198-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/30/2015] [Indexed: 12/13/2022] Open
Abstract
The progression of colorectal cancer is commonly characterized by accumulation of genetic or epigenetic abnormalities, altering regulation of gene expression as well as normal protein structures and functions. Nonetheless, there are some questions that remain to be elucidated, such as the origin of cancer cells and populations of cells initiating and propagating tumor development. Currently, there are two rival theories describing the process of carcinogenesis. One is the stochastic model, arguing that any cell is capable of initiating and triggering the development of cancer. Meanwhile, the cancer stem cell model hypothesizes that only a small fraction of stem cells possesses cancer-promoting properties. Typically, colorectal cancer stem cells (CSCs) share the same molecular signaling profiles with normal stem cells or embryonic stem cells, such as Wnt, Notch, TGF-β, and Hedgehog. Nevertheless, CSCs differ from normal stem cells and the bulk of tumor cells in their tumorigenic potential and susceptibility to chemotherapeutic drugs. This may be a possible explanation of the high percentage of cancer recurrence in patients who underwent chemotherapeutic treatment and surgery. This review article focuses on the colorectal cancer stem cell biomarkers and the role of upregulated signaling pathways implicated in the initiation and progression of colorectal cancer.
Collapse
Affiliation(s)
- Danysh Abetov
- Department of Regenerative Medicine and Artificial Organs, Centre for Life Sciences, Nazarbayev University, Unit 9, 53 Kabanbay batyr Ave., Astana, Kazakhstan, 010000
| | | | | | | |
Collapse
|
582
|
Combined QTL and selective sweep mappings with coding SNP annotation and cis-eQTL analysis revealed PARK2 and JAG2 as new candidate genes for adiposity regulation. G3-GENES GENOMES GENETICS 2015; 5:517-29. [PMID: 25653314 PMCID: PMC4390568 DOI: 10.1534/g3.115.016865] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Very few causal genes have been identified by quantitative trait loci (QTL) mapping because of the large size of QTL, and most of them were identified thanks to functional links already known with the targeted phenotype. Here, we propose to combine selection signature detection, coding SNP annotation, and cis-expression QTL analyses to identify potential causal genes underlying QTL identified in divergent line designs. As a model, we chose experimental chicken lines divergently selected for only one trait, the abdominal fat weight, in which several QTL were previously mapped. Using new haplotype-based statistics exploiting the very high SNP density generated through whole-genome resequencing, we found 129 significant selective sweeps. Most of the QTL colocalized with at least one sweep, which markedly narrowed candidate region size. Some of those sweeps contained only one gene, therefore making them strong positional causal candidates with no presupposed function. We then focused on two of these QTL/sweeps. The absence of nonsynonymous SNPs in their coding regions strongly suggests the existence of causal mutations acting in cis on their expression, confirmed by cis-eQTL identification using either allele-specific expression or genetic mapping analyses. Additional expression analyses of those two genes in the chicken and mice contrasted for adiposity reinforces their link with this phenotype. This study shows for the first time the interest of combining selective sweeps mapping, coding SNP annotation and cis-eQTL analyses for identifying causative genes for a complex trait, in the context of divergent lines selected for this specific trait. Moreover, it highlights two genes, JAG2 and PARK2, as new potential negative and positive key regulators of adiposity in chicken and mice.
Collapse
|
583
|
Lindström NO, Lawrence ML, Burn SF, Johansson JA, Bakker ERM, Ridgway RA, Chang CH, Karolak MJ, Oxburgh L, Headon DJ, Sansom OJ, Smits R, Davies JA, Hohenstein P. Integrated β-catenin, BMP, PTEN, and Notch signalling patterns the nephron. eLife 2015; 3:e04000. [PMID: 25647637 PMCID: PMC4337611 DOI: 10.7554/elife.04000] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 12/28/2014] [Indexed: 12/13/2022] Open
Abstract
The different segments of the nephron and glomerulus in the kidney balance the processes of water homeostasis, solute recovery, blood filtration, and metabolite excretion. When segment function is disrupted, a range of pathological features are presented. Little is known about nephron patterning during embryogenesis. In this study, we demonstrate that the early nephron is patterned by a gradient in β-catenin activity along the axis of the nephron tubule. By modifying β-catenin activity, we force cells within nephrons to differentiate according to the imposed β-catenin activity level, thereby causing spatial shifts in nephron segments. The β-catenin signalling gradient interacts with the BMP pathway which, through PTEN/PI3K/AKT signalling, antagonises β-catenin activity and promotes segment identities associated with low β-catenin activity. β-catenin activity and PI3K signalling also integrate with Notch signalling to control segmentation: modulating β-catenin activity or PI3K rescues segment identities normally lost by inhibition of Notch. Our data therefore identifies a molecular network for nephron patterning.
Collapse
Affiliation(s)
- Nils O Lindström
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Melanie L Lawrence
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Sally F Burn
- Department of Genetics and Development, Columbia University, New York, United States
| | - Jeanette A Johansson
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Elvira RM Bakker
- Laboratory of Gastroenterology and Hepatology, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Rachel A Ridgway
- Department of Invasion and Metastasis, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - C-Hong Chang
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Michele J Karolak
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Denis J Headon
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Owen J Sansom
- Beatston Institute for Cancer Research, Glasgow, United Kingdom
| | - Ron Smits
- Laboratory of Gastroenterology and Hepatology, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Jamie A Davies
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Hohenstein
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| |
Collapse
|
584
|
Barcena Menendez D, Senthivel VR, Isalan M. Sender-receiver systems and applying information theory for quantitative synthetic biology. Curr Opin Biotechnol 2015; 31:101-7. [PMID: 25282688 PMCID: PMC4332572 DOI: 10.1016/j.copbio.2014.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/21/2014] [Indexed: 12/31/2022]
Abstract
Sender-receiver (S-R) systems abound in biology, with communication systems sending information in various forms. Information theory provides a quantitative basis for analysing these processes and is being applied to study natural genetic, enzymatic and neural networks. Recent advances in synthetic biology are providing us with a wealth of artificial S-R systems, giving us quantitative control over networks with a finite number of well-characterised components. Combining the two approaches can help to predict how to maximise signalling robustness, and will allow us to make increasingly complex biological computers. Ultimately, pushing the boundaries of synthetic biology will require moving beyond engineering the flow of information and towards building more sophisticated circuits that interpret biological meaning.
Collapse
Affiliation(s)
- Diego Barcena Menendez
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Vivek Raj Senthivel
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Mark Isalan
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
585
|
Pfeuty B. A computational model for the coordination of neural progenitor self-renewal and differentiation through Hes1 dynamics. Development 2015; 142:477-85. [DOI: 10.1242/dev.112649] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Proper tissue development requires that stem/progenitor cells precisely coordinate cell division and differentiation in space and time. Notch-Hes1 intercellular signaling, which affects both differentiation and cell cycle progression and directs cell fate decisions at various developmental stages in many cell types, is central to this process. This study explored whether the pattern of connections among the cell cycle regulatory module, the Notch effector Hes1 and the proneural factor Ngn2 could explain salient aspects of cell fate determination in neural progenitors. A mathematical model that includes mutual interactions between Hes1, Ngn2 and G1-phase regulators was constructed and simulated at the single- and two-cell levels. By differentially regulating G1-phase progression, Hes1 and Ngn2 are shown to induce two contrasting cell cycle arrest states in early and late G1, respectively. Indeed, steady Hes1 overexpression promotes reversible quiescence by downregulating activators of G0/G1 exit and Ngn2. Ngn2 also downregulates activators of G0/G1 exit, but cooperates with Cip/Kip proteins to prevent G1/S transit, whereby it promotes G1-phase lengthening and, ultimately, contributes to reinforcing an irreversible late G1 arrest coincident with terminal differentiation. In this scheme, Hes1 oscillation in single cells is able to maintain a labile proliferation state in dynamic balance with two competing cell fate outputs associated with Hes1-mediated and Ngn2-mediated cell cycle arrest states. In Delta/Notch-connected cells, Hes1 oscillations and a lateral inhibition mechanism combine to establish heterogeneous Hes1, Ngn2 and cell cycle dynamics between proliferating neural progenitors, thereby increasing the chances of asymmetric cell fate decisions and improving the reliability of commitment to differentiation.
Collapse
Affiliation(s)
- Benjamin Pfeuty
- CNRS, UMR 8523, Université de Lille USTL, Villeneuve d'Ascq 59650, France
| |
Collapse
|
586
|
Jagged-Delta asymmetry in Notch signaling can give rise to a Sender/Receiver hybrid phenotype. Proc Natl Acad Sci U S A 2015; 112:E402-9. [PMID: 25605936 DOI: 10.1073/pnas.1416287112] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Notch signaling pathway mediates cell-fate determination during embryonic development, wound healing, and tumorigenesis. This pathway is activated when the ligand Delta or the ligand Jagged of one cell interacts with the Notch receptor of its neighboring cell, releasing the Notch Intracellular Domain (NICD) that activates many downstream target genes. NICD affects ligand production asymmetrically--it represses Delta, but activates Jagged. Although the dynamical role of Notch-Jagged signaling remains elusive, it is widely recognized that Notch-Delta signaling behaves as an intercellular toggle switch, giving rise to two distinct fates that neighboring cells adopt--Sender (high ligand, low receptor) and Receiver (low ligand, high receptor). Here, we devise a specific theoretical framework that incorporates both Delta and Jagged in Notch signaling circuit to explore the functional role of Jagged in cell-fate determination. We find that the asymmetric effect of NICD renders the circuit to behave as a three-way switch, giving rise to an additional state--a hybrid Sender/Receiver (medium ligand, medium receptor). This phenotype allows neighboring cells to both send and receive signals, thereby attaining similar fates. We also show that due to the asymmetric effect of the glycosyltransferase Fringe, different outcomes are generated depending on which ligand is dominant: Delta-mediated signaling drives neighboring cells to have an opposite fate; Jagged-mediated signaling drives the cell to maintain a similar fate to that of its neighbor. We elucidate the role of Jagged in cell-fate determination and discuss its possible implications in understanding tumor-stroma cross-talk, which frequently entails Notch-Jagged communication.
Collapse
|
587
|
Sriram R, Lo V, Pryce B, Antonova L, Mears AJ, Daneshmand M, McKay B, Conway SJ, Muller WJ, Sabourin LA. Loss of periostin/OSF-2 in ErbB2/Neu-driven tumors results in androgen receptor-positive molecular apocrine-like tumors with reduced Notch1 activity. Breast Cancer Res 2015; 17:7. [PMID: 25592291 PMCID: PMC4355979 DOI: 10.1186/s13058-014-0513-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Periostin (Postn) is a secreted cell adhesion protein that activates signaling pathways to promote cancer cell survival, angiogenesis, invasion, and metastasis. Interestingly, Postn is frequently overexpressed in numerous human cancers, including breast, lung, colon, pancreatic, and ovarian cancer. METHODS Using transgenic mice expressing the Neu oncogene in the mammary epithelium crossed into Postn-deficient animals, we have assessed the effect of Postn gene deletion on Neu-driven mammary tumorigenesis. RESULTS Although Postn is exclusively expressed in the stromal fibroblasts of the mammary gland, Postn deletion does not affect mammary gland outgrowth during development or pregnancy. Furthermore, we find that loss of Postn in the mammary epithelium does not alter breast tumor initiation or growth in mouse mammary tumor virus (MMTV)-Neu expressing mice but results in an apocrine-like tumor phenotype. Surprisingly, we find that tumors derived from Postn-null animals express low levels of Notch protein and Hey1 mRNA but increased expression of androgen receptor (AR) and AR target genes. We show that tumor cells derived from wild-type animals do not proliferate when transplanted in a Postn-null environment but that this growth defect is rescued by the overexpression of active Notch or the AR target gene prolactin-induced protein (PIP/GCDFP-15). CONCLUSIONS Together our data suggest that loss of Postn in an ErbB2/Neu/HER2 overexpression model results in apocrine-like tumors that activate an AR-dependent pathway. This may have important implications for the treatment of breast cancers involving the therapeutic targeting of periostin or Notch signaling.
Collapse
Affiliation(s)
- Roshan Sriram
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Vivian Lo
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Benjamin Pryce
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Lilia Antonova
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Alan J Mears
- Children's Hospital of Eastern Ontario, Research Institute, 501 Smyth Road, Ottawa, ON, K1H8L6, Canada.
| | - Manijeh Daneshmand
- Ottawa Hospital Research Institute, Cancer Therapeutics, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| | - Bruce McKay
- Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, 705 Riley Hospital Drive, Indianapolis, IN, 46202, USA.
| | - William J Muller
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, 1200 Pine Avenue West, Montreal, QC, H3G 1A1, Canada.
| | - Luc A Sabourin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada. .,Ottawa Hospital Research Institute, Cancer Therapeutics, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
588
|
Wang K, Zhang Q, Li D, Ching K, Zhang C, Zheng X, Ozeck M, Shi S, Li X, Wang H, Rejto P, Christensen J, Olson P. PEST domain mutations in Notch receptors comprise an oncogenic driver segment in triple-negative breast cancer sensitive to a γ-secretase inhibitor. Clin Cancer Res 2015; 21:1487-96. [PMID: 25564152 DOI: 10.1158/1078-0432.ccr-14-1348] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To identify and characterize novel, activating mutations in Notch receptors in breast cancer and to determine response to the gamma secretase inhibitor (GSI) PF-03084014. EXPERIMENTAL DESIGN We used several computational approaches, including novel algorithms, to analyze next-generation sequencing data and related omic datasets from The Cancer Genome Atlas (TCGA) breast cancer cohort. Patient-derived xenograft (PDX) models were sequenced, and Notch-mutant models were treated with PF-03084014. Gene-expression and functional analyses were performed to study the mechanism of activation through mutation and inhibition by PF-03084014. RESULTS We identified mutations within and upstream of the PEST domains of NOTCH1, NOTCH2, and NOTCH3 in the TCGA dataset. Mutations occurred via several genetic mechanisms and compromised the function of the PEST domain, a negative regulatory domain commonly mutated in other cancers. Focal amplifications of NOTCH2 and NOTCH3 were also observed, as were heterodimerization or extracellular domain mutations at lower incidence. Mutations and amplifications often activated the Notch pathway as evidenced by increased expression of canonical Notch target genes, and functional mutations were significantly enriched in the triple-negative breast cancer subtype (TNBC). PDX models were also identified that harbored PEST domain mutations, and these models were highly sensitive to PF-03084014. CONCLUSIONS This work suggests that Notch-altered breast cancer constitutes a bona fide oncogenic driver segment with the most common alteration being PEST domain mutations present in multiple Notch receptors. Importantly, functional studies suggest that this newly identified class can be targeted with Notch inhibitors, including GSIs.
Collapse
Affiliation(s)
- Kai Wang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Qin Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Danan Li
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Keith Ching
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Cathy Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Xianxian Zheng
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Mark Ozeck
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Stephanie Shi
- External Research Solutions, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Xiaorong Li
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Hui Wang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Paul Rejto
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - James Christensen
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Peter Olson
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California.
| |
Collapse
|
589
|
Adamska M. Developmental Signalling and Emergence of Animal Multicellularity. EVOLUTIONARY TRANSITIONS TO MULTICELLULAR LIFE 2015. [DOI: 10.1007/978-94-017-9642-2_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
590
|
NOTCH1 and FBXW7 mutations favor better outcome in pediatric South Indian T-cell acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2015; 37:e23-30. [PMID: 25493453 DOI: 10.1097/mph.0000000000000290] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The NOTCH1 signaling pathway is essential for hematopoiesis and a critical regulatory step for T-cell proliferation and maturation. The E3 ubiquitin ligase FBXW7 controls NOTCH1 protein stability. Mutations in NOTCH1/FBXW7 activate NOTCH signaling and are of prognostic significance in patients with T-cell acute lymphoblastic leukemia (T-ALL). In this study we analyzed NOTCH1 and FBXW7 mutations in 50 South Indian T-ALL patients treated by a modified ALL BFM 95 regimen. The hot spot exons (HD-N, HD-C, TAD, and PEST) of NOTCH1 and exons 9 of the 10 of FBXW7 were polymerase chain reaction amplified and sequenced. In total, 20 of the 50 (40%) T-ALL patients revealed heterozygous mutations in the NOTCH1 domains, and a predominance of missense mutations in HD-N (70%) and PEST (15%) domains. FBXW7 mutations were detected in 5 of the 50 (10%) T-ALL patients. T-ALL patients with NOTCH1/FBXW7 mutations expressed higher protein level of NOTCH1 compared with patients without NOTCH1/FBXW7 mutations. Six of the mutations detected in NOTCH1 were not reported previously. When tested in a Dual Luciferase Renilla reporter assay some of these conferred increased NOTCH activity, suggesting that these are activating mutations. Importantly, 13 of the 20 (65%) NOTCH1/FBXW7-mutated T-ALL patients showed a good prednisone response (P=0.01) and a better clinical outcome compared with NOTCH1/FBXW7 nonmutated patients (P=0.03). These data suggest that NOTCH1/FBXW7 mutations are present in T-ALL patients from Southern India and may be useful biomarkers to predict prognosis in T-ALL.
Collapse
|
591
|
Kalucka J, Missiaen R, Georgiadou M, Schoors S, Lange C, De Bock K, Dewerchin M, Carmeliet P. Metabolic control of the cell cycle. Cell Cycle 2015; 14:3379-88. [PMID: 26431254 PMCID: PMC4825590 DOI: 10.1080/15384101.2015.1090068] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 08/29/2015] [Indexed: 12/14/2022] Open
Abstract
Cell division is a metabolically demanding process, requiring the production of large amounts of energy and biomass. Not surprisingly therefore, a cell's decision to initiate division is co-determined by its metabolic status and the availability of nutrients. Emerging evidence reveals that metabolism is not only undergoing substantial changes during the cell cycle, but it is becoming equally clear that metabolism regulates cell cycle progression. Here, we overview the emerging role of those metabolic pathways that have been best characterized to change during or influence cell cycle progression. We then studied how Notch signaling, a key angiogenic pathway that inhibits endothelial cell (EC) proliferation, controls EC metabolism (glycolysis) during the cell cycle.
Collapse
Affiliation(s)
- Joanna Kalucka
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| | - Rindert Missiaen
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| | - Maria Georgiadou
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
- Present address: Turku Centre for Biotechnology; Turku, Finland
| | - Sandra Schoors
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| | - Christian Lange
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
- Present address: Exercise Physiology Research Group; Department of Kinesiology; KU Leuven; Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link; Department of Oncology; KU Leuven; Leuven, Belgium
- Laboratory of Angiogenesis and Neurovascular link; Vesalius Research Center; VIB, Leuven, Belgium
| |
Collapse
|
592
|
|
593
|
Geisler F, Strazzabosco M. Emerging roles of Notch signaling in liver disease. Hepatology 2015; 61:382-92. [PMID: 24930574 PMCID: PMC4268103 DOI: 10.1002/hep.27268] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/11/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
This review critically discusses the most recent advances in the role of Notch signaling in liver development, homeostasis, and disease. It is now clear that the significance of Notch in determining mammalian cell fates and functions extends beyond development, and Notch is a major regular of organ homeostasis. Moreover, Notch signaling is reactivated upon injury and regulates the complex interactions between the distinct liver cell types involved in the repair process. Notch is also involved in the regulation of liver metabolism, inflammation, and cancer. The net effects of Notch signaling are highly variable and finely regulated at multiple levels, but also depend on the specific cellular context in which Notch is activated. Persistent activation of Notch signaling is associated with liver malignancies, such as hepatocellular carcinoma with stem cell features and intrahepatic cholangiocarcinoma. The complexity of the pathway provides several possible targets for agents able to inhibit Notch. However, further cell- and context-specific in-depth understanding of Notch signaling in liver homeostasis and disease will be essential to translate these concepts into clinical practice and be able to predict benefits and risks of evolving therapies.
Collapse
Affiliation(s)
- Fabian Geisler
- 2nd Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Mario Strazzabosco
- Liver Center & Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA,Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
594
|
Abstract
The use of umbilical cord blood (UCB) as an alternative haematopoietic cell source in lieu of bone marrow for haematopoietic reconstitution is increasingly becoming a mainstay treatment for both malignant and nonmalignant diseases, as most individuals will have at least one available, suitably HLA-matched unit of blood. The principal limitation of UCB is the low and finite number of haematopoietic stem and progenitor cells (HSPC) relative to the number found in a typical bone marrow or mobilized peripheral blood allograft, which leads to prolonged engraftment times. In an attempt to overcome this obstacle, strategies that are often based on native processes occurring in the bone marrow microenvironment or 'niche' have been developed with the goal of accelerating UCB engraftment. In broad terms, the two main approaches have been either to expand UCB HSPC ex vivo before transplantation, or to modulate HSPC functionality to increase the efficiency of HSPC homing to the bone marrow niche after transplant both of which enhance the biological activities of the engrafted HSPC. Several early phase clinical trials of these approaches have reported promising results.
Collapse
|
595
|
Zhang X, Li Y, Xu H, Zhang YW. The γ-secretase complex: from structure to function. Front Cell Neurosci 2014; 8:427. [PMID: 25565961 PMCID: PMC4263104 DOI: 10.3389/fncel.2014.00427] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
One of the most critical pathological features of Alzheimer’s disease (AD) is the accumulation of β-amyloid (Aβ) peptides that form extracellular senile plaques in the brain. Aβ is derived from β-amyloid precursor protein (APP) through sequential cleavage by β- and γ-secretases. γ-secretase is a high molecular weight complex minimally composed of four components: presenilins (PS), nicastrin, anterior pharynx defective 1 (APH-1), and presenilin enhancer 2 (PEN-2). In addition to APP, γ-secretase also cleaves many other type I transmembrane (TM) protein substrates. As a crucial enzyme for Aβ production, γ-secretase is an appealing therapeutic target for AD. Here, we summarize current knowledge on the structure and function of γ-secretase, as well as recent progress in developing γ-secretase targeting drugs for AD treatment.
Collapse
Affiliation(s)
- Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China ; Degenerative Disease Research Program, Sanford-Burnham Medical Research Institute La Jolla, CA, USA
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| |
Collapse
|
596
|
Henshall TL, Keller A, He L, Johansson BR, Wallgard E, Raschperger E, Mäe MA, Jin S, Betsholtz C, Lendahl U. Notch3 is necessary for blood vessel integrity in the central nervous system. Arterioscler Thromb Vasc Biol 2014; 35:409-20. [PMID: 25477343 DOI: 10.1161/atvbaha.114.304849] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMC) are important for contraction, blood flow distribution, and regulation of blood vessel diameter, but to what extent they contribute to the integrity of blood vessels and blood-brain barrier function is less well understood. In this report, we explored the impact of the loss of VSMC in the Notch3(-/-) mouse on blood vessel integrity in the central nervous system. APPROACH AND RESULTS Notch3(-/-) mice showed focal disruptions of the blood-brain barrier demonstrated by extravasation of tracers accompanied by fibrin deposition in the retinal vasculature. This blood-brain barrier leakage was accompanied by a regionalized and patchy loss of VSMC, with VSMC gaps predominantly in arterial resistance vessels of larger caliber. The loss of VSMC appeared to be caused by progressive degeneration of VSMC resulting in a gradual loss of VSMC marker expression and a progressive acquisition of an aberrant VSMC phenotype closer to the gaps, followed by enhanced apoptosis and cellular disintegration in the gaps. Arterial VSMC were the only mural cell type that was morphologically affected, despite Notch3 also being expressed in pericytes. Transcriptome analysis of isolated brain microvessels revealed gene expression changes in Notch3(-/-) mice consistent with loss of arterial VSMC and presumably secondary transcriptional changes were observed in endothelial genes, which may explain the compromised vascular integrity. CONCLUSIONS We demonstrate that Notch3 is important for survival of VSMC, and reveal a critical role for Notch3 and VSMC in blood vessel integrity and blood-brain barrier function in the mammalian vasculature.
Collapse
Affiliation(s)
- Tanya L Henshall
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Annika Keller
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Liqun He
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Bengt R Johansson
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Elisabet Wallgard
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Elisabeth Raschperger
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Maarja Andaloussi Mäe
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Shaobo Jin
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.)
| | - Christer Betsholtz
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.).
| | - Urban Lendahl
- From the Department of Cell and Molecular Biology (T.H., S.J., U.L.) and Department of Medical Biochemistry and Biophysics, Division of Vascular Biology (C.B., E.R.), Karolinska Institute, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden (A.K., L.H., E.R., M.A.M., C.B.); EM Unit, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden (B.R.J.); and Octapharma AB, Stockholm, Sweden (E.W.).
| |
Collapse
|
597
|
Hallaq R, Volpicelli F, Cuchillo-Ibanez I, Hooper C, Mizuno K, Uwanogho D, Causevic M, Asuni A, To A, Soriano S, Giese KP, Lovestone S, Killick R. The Notch intracellular domain represses CRE-dependent transcription. Cell Signal 2014; 27:621-9. [PMID: 25479589 DOI: 10.1016/j.cellsig.2014.11.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/27/2014] [Accepted: 11/27/2014] [Indexed: 12/25/2022]
Abstract
Members of the cyclic-AMP response-element binding protein (CREB) transcription factor family regulate the expression of genes needed for long-term memory formation. Loss of Notch impairs long-term, but not short-term, memory in flies and mammals. We investigated if the Notch-1 (N1) exerts an effect on CREB-dependent gene transcription. We observed that N1 inhibits CREB mediated activation of cyclic-AMP response element (CRE) containing promoters in a γ-secretase-dependent manner. We went on to find that the γ-cleaved N1 intracellular domain (N1ICD) sequesters nuclear CREB1α, inhibits cAMP/PKA-mediated neurite outgrowth and represses the expression of specific CREB regulated genes associated with learning and memory in primary cortical neurons. Similar transcriptional effects were observed with the N2ICD, N3ICD and N4ICDs. Together, these observations indicate that the effects of Notch on learning and memory are, at least in part, via an effect on CREB-regulated gene expression.
Collapse
Affiliation(s)
- Rania Hallaq
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Floriana Volpicelli
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Inmaculada Cuchillo-Ibanez
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Claudie Hooper
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Keiko Mizuno
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Dafe Uwanogho
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Mirsada Causevic
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Ayodeji Asuni
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Alvina To
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Salvador Soriano
- Department of Anatomy, Loma Linda University School of Medicine, Loma Linda, Evans Hall B08, 24785 Stewart Street, Loma Linda, CA 92354, USA
| | - K Peter Giese
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | - Simon Lovestone
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford OX3 7JX, UK
| | - Richard Killick
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, Denmark Hill, London SE5 8AF, UK.
| |
Collapse
|
598
|
Cast AE, Walter TJ, Huppert SS. Vascular patterning sets the stage for macro and micro hepatic architecture. Dev Dyn 2014; 244:497-506. [PMID: 25370311 DOI: 10.1002/dvdy.24222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 01/20/2023] Open
Abstract
Background The liver is a complex organ with a variety of tissue components that require a precise architecture for optimal function of metabolic and detoxification processes. As a result of the delicate orchestration required between the various hepatic tissues, it is not surprising that impairment of hepatic function can be caused by a variety of factors leading to chronic liver disease. Results Despite the growing rate of chronic liver disease, there are currently few effective treatment options besides orthotopic liver transplantation. Better therapeutic options reside in the potential for genetic and cellular therapies that promote progenitor cell activation aiding de novo epithelial and vascular regeneration, cell replacement, or population of bioartificial hepatic devices. In order to explore this area of new therapeutic potential, it is crucial to understand the factors that promote hepatic function through regulating cell identities and tissue architecture. Conclusions In this commentary, we review the signals regulating liver cell fates during development and regeneration and highlight the importance of patterning the hepatic vascular systems to set the groundwork for the macro and micro hepatic architecture of the epithelium.
Collapse
Affiliation(s)
- Ashley E Cast
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | | |
Collapse
|
599
|
Rivers OS, Videau P, Callahan SM. Mutation of
sepJ
reduces the intercellular signal range of a
hetN
‐dependent paracrine signal, but not of a
patS
‐dependent signal, in the filamentous cyanobacterium
A
nabaena
sp. strain
PCC
7120. Mol Microbiol 2014; 94:1260-71. [DOI: 10.1111/mmi.12836] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Orion S. Rivers
- Department of MicrobiologyUniversity of Hawai'i at Mānoa Honolulu HI 96822 USA
| | - Patrick Videau
- Department of MicrobiologyUniversity of Hawai'i at Mānoa Honolulu HI 96822 USA
| | - Sean M. Callahan
- Department of MicrobiologyUniversity of Hawai'i at Mānoa Honolulu HI 96822 USA
| |
Collapse
|
600
|
Manning CS, Hooper S, Sahai EA. Intravital imaging of SRF and Notch signalling identifies a key role for EZH2 in invasive melanoma cells. Oncogene 2014; 34:4320-32. [PMID: 25381824 PMCID: PMC4349503 DOI: 10.1038/onc.2014.362] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 07/01/2014] [Accepted: 07/24/2014] [Indexed: 12/18/2022]
Abstract
The acquisition of cell motility is an early step in melanoma metastasis. Here we use intravital imaging of signalling reporter cell-lines combined with genome-wide transcriptional analysis to define signalling pathways and genes associated with melanoma metastasis. Intravital imaging revealed heterogeneous cell behaviour in vivo: <10% of cells were motile and both singly moving cells and streams of cells were observed. Motile melanoma cells had increased Notch- and SRF-dependent transcription. Subsequent genome-wide analysis identified an overlapping set of genes associated with high Notch and SRF activity. We identified EZH2, a histone methyltransferase in the Polycomb repressive complex 2, as a regulator of these genes. Heterogeneity of EZH2 levels is observed in melanoma models, and co-ordinated upregulation of genes positively regulated by EZH2 is associated with melanoma metastasis. EZH2 was also identified as regulating the amelanotic phenotype of motile cells in vivo by suppressing expression of the P-glycoprotein Oca2. Analysis of patient samples confirmed an inverse relationship between EZH2 levels and pigment. EZH2 targeting with siRNA and chemical inhibition reduced invasion in mouse and human melanoma cell lines. The EZH2-regulated genes KIF2C and KIF22 are required for melanoma cell invasion and important for lung colonization. We propose that heterogeneity in EZH2 levels leads to heterogeneous expression of a cohort of genes associated with motile behaviour including KIF2C and KIF22. EZH2-dependent increased expression of these genes promotes melanoma cell motility and early steps in metastasis.
Collapse
Affiliation(s)
- C S Manning
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK
| | - S Hooper
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK
| | - E A Sahai
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, London, UK
| |
Collapse
|