51
|
Takam Kamga P, Bazzoni R, Dal Collo G, Cassaro A, Tanasi I, Russignan A, Tecchio C, Krampera M. The Role of Notch and Wnt Signaling in MSC Communication in Normal and Leukemic Bone Marrow Niche. Front Cell Dev Biol 2021; 8:599276. [PMID: 33490067 PMCID: PMC7820188 DOI: 10.3389/fcell.2020.599276] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Notch and Wnt signaling are highly conserved intercellular communication pathways involved in developmental processes, such as hematopoiesis. Even though data from literature support a role for these two pathways in both physiological hematopoiesis and leukemia, there are still many controversies concerning the nature of their contribution. Early studies, strengthened by findings from T-cell acute lymphoblastic leukemia (T-ALL), have focused their investigation on the mutations in genes encoding for components of the pathways, with limited results except for B-cell chronic lymphocytic leukemia (CLL); in because in other leukemia the two pathways could be hyper-expressed without genetic abnormalities. As normal and malignant hematopoiesis require close and complex interactions between hematopoietic cells and specialized bone marrow (BM) niche cells, recent studies have focused on the role of Notch and Wnt signaling in the context of normal crosstalk between hematopoietic/leukemia cells and stromal components. Amongst the latter, mesenchymal stromal/stem cells (MSCs) play a pivotal role as multipotent non-hematopoietic cells capable of giving rise to most of the BM niche stromal cells, including fibroblasts, adipocytes, and osteocytes. Indeed, MSCs express and secrete a broad pattern of bioactive molecules, including Notch and Wnt molecules, that support all the phases of the hematopoiesis, including self-renewal, proliferation and differentiation. Herein, we provide an overview on recent advances on the contribution of MSC-derived Notch and Wnt signaling to hematopoiesis and leukemia development.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giada Dal Collo
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Adriana Cassaro
- Hematology Unit, Department of Oncology, Niguarda Hospital, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Anna Russignan
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
52
|
Vlachakis D, Papageorgiou L, Papadaki A, Georga M, Kossida S, Eliopoulos E. An updated evolutionary study of the Notch family reveals a new ancient origin and novel invariable motifs as potential pharmacological targets. PeerJ 2020; 8:e10334. [PMID: 33194454 PMCID: PMC7649014 DOI: 10.7717/peerj.10334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/19/2020] [Indexed: 01/02/2023] Open
Abstract
Notch family proteins play a key role in a variety of developmental processes by controlling cell fate decisions and operating in a great number of biological processes in several organ systems, such as hematopoiesis, somatogenesis, vasculogenesis, neurogenesis and homeostasis. The Notch signaling pathway is crucial for the majority of developmental programs and regulates multiple pathogenic processes. Notch family receptors' activation has been largely related to its multiple effects in sustaining oncogenesis. The Notch signaling pathway constitutes an ancient and conserved mechanism for cell to cell communication. Much of what is known about Notch family proteins function comes from studies done in Caenorhabditis Elegans and Drosophila Melanogaster. Although, human Notch homologs had also been identified, the molecular mechanisms which modulate the Notch signaling pathway remained substantially unknown. In this study, an updated evolutionary analysis of the Notch family members among 603 different organisms of all kingdoms, from bacteria to humans, was performed in order to discover key regions that have been conserved throughout evolution and play a major role in the Notch signaling pathway. The major goal of this study is the presentation of a novel updated phylogenetic tree for the Notch family as a reliable phylogeny "map", in order to correlate information of the closely related members and identify new possible pharmacological targets that can be used in pathogenic cases, including cancer.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Louis Papageorgiou
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, Athens, Greece
| | - Ariadne Papadaki
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Maria Georga
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Sofia Kossida
- IMGT, The International ImMunoGeneTics Information System, Université de Montpellier, Laboratoire d’ImmunoGénétique Moléculaire and Institut de Génétique Humaine, University of Montpellier, Montpellier, France
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
53
|
Notch Pathway: A Journey from Notching Phenotypes to Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1287:201-222. [PMID: 33034034 DOI: 10.1007/978-3-030-55031-8_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Notch is a key evolutionary conserved pathway, which has fascinated and engaged the work of investigators in an uncountable number of biological fields, from development of metazoans to immunotherapy for cancer. The study of Notch has greatly contributed to the understanding of cancer biology and a substantial effort has been spent in designing Notch-targeting therapies. Due to its broad involvement in cancer, targeting Notch would allow to virtually modulate any aspect of the disease. However, this means that Notch-based therapies must be highly specific to avoid off-target effects. This review will present the newest mechanistic and therapeutic advances in the Notch field and discuss the promises and challenges of this constantly evolving field.
Collapse
|
54
|
Zohorsky K, Mequanint K. Designing Biomaterials to Modulate Notch Signaling in Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:383-410. [PMID: 33040694 DOI: 10.1089/ten.teb.2020.0182] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The design of cell-instructive biomaterials for tissue engineering and regenerative medicine is at a crossroads. Although the conventional tissue engineering approach is top-down (cells seeded to macroporous scaffolds and mature to form tissues), bottom-up tissue engineering strategies are becoming appealing. With such developments, we can study cell signaling events, thus enabling functional tissue assembly in physiologic and diseased models. Among many important signaling pathways, the Notch signaling pathway is the most diverse in its influence during tissue morphogenesis and repair following injury. Although Notch signaling is extensively studied in developmental biology and cancer biology, our knowledge of designing biomaterial-based Notch signaling platforms and incorporating Notch signaling components into engineered tissue systems is limited. By incorporating Notch signaling to tissue engineering scaffolds, we can direct cell-specific responses and improve engineered tissue maturation. This review will discuss recent progress in the development of Notch signaling biomaterials as a promising target to control cellular fate decisions, including the influences of ligand identity, biophysical material cues, ligand presentation strategies, and mechanotransduction. Notch signaling is consequently of interest to direct, control, and reprogram cellular behavior on a biomaterial surface. We anticipate that discussions in this article will allow for enhanced knowledge and insight into designing Notch targeted biomaterials for various tissue engineering and cell fate determinations. Impact statement Notch signaling is recognized as an important pathway in tissue engineering and regenerative medicine; however, there is no systematic review on this topic. The comprehensive review and perspectives presented here provide an in-depth discussion on ligand presentation strategies both in 2D and in 3D cell culture environments involving biomaterials/scaffolds. In addition, this review article provides insight into the challenges in designing cell surrogate biomaterials capable of providing Notch signals. To the best of the authors' knowledge, this is the first review relevant to the fields of tissue engineering.
Collapse
Affiliation(s)
- Kathleen Zohorsky
- School of Biomedical Engineering and The University of Western Ontario, London, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering and The University of Western Ontario, London, Canada.,Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| |
Collapse
|
55
|
Kobia FM, Preusse K, Dai Q, Weaver N, Hass MR, Chaturvedi P, Stein SJ, Pear WS, Yuan Z, Kovall RA, Kuang Y, Eafergen N, Sprinzak D, Gebelein B, Brunskill EW, Kopan R. Notch dimerization and gene dosage are important for normal heart development, intestinal stem cell maintenance, and splenic marginal zone B-cell homeostasis during mite infestation. PLoS Biol 2020; 18:e3000850. [PMID: 33017398 PMCID: PMC7561103 DOI: 10.1371/journal.pbio.3000850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/15/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cooperative DNA binding is a key feature of transcriptional regulation. Here we examined the role of cooperativity in Notch signaling by CRISPR-mediated engineering of mice in which neither Notch1 nor Notch2 can homo- or heterodimerize, essential for cooperative binding to sequence-paired sites (SPS) located near many Notch-regulated genes. Although most known Notch-dependent phenotypes were unaffected in Notch1/2 dimer-deficient mice, a subset of tissues proved highly sensitive to loss of cooperativity. These phenotypes include heart development, compromised viability in combination with low gene dose, and the gut, developing ulcerative colitis in response to 1% dextran sulfate sodium (DSS). The most striking phenotypes-gender imbalance and splenic marginal zone B-cell lymphoma-emerged in combination with gene dose reduction or when challenged by chronic fur mite infestation. This study highlights the role of the environment in malignancy and colitis and is consistent with Notch-dependent anti-parasite immune responses being compromised in Notch dimer-deficient animals.
Collapse
Affiliation(s)
- Francis M. Kobia
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kristina Preusse
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Quanhui Dai
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Nicholas Weaver
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Matthew R. Hass
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Praneet Chaturvedi
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Sarah J. Stein
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Warren S. Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Zhenyu Yuan
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rhett A. Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Yi Kuang
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Natanel Eafergen
- School of Neurobiology, Biochemistry, and Biophysics, The George S. Wise Faculty of Life Sciences Tel Aviv University, Tel Aviv, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry, and Biophysics, The George S. Wise Faculty of Life Sciences Tel Aviv University, Tel Aviv, Israel
| | - Brian Gebelein
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Eric W. Brunskill
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| |
Collapse
|
56
|
Manshaei R, Merico D, Reuter MS, Engchuan W, Mojarad BA, Chaturvedi R, Heung T, Pellecchia G, Zarrei M, Nalpathamkalam T, Khan R, Okello JBA, Liston E, Curtis M, Yuen RKC, Marshall CR, Jobling RK, Oechslin E, Wald RM, Silversides CK, Scherer SW, Kim RH, Bassett AS. Genes and Pathways Implicated in Tetralogy of Fallot Revealed by Ultra-Rare Variant Burden Analysis in 231 Genome Sequences. Front Genet 2020; 11:957. [PMID: 33110418 PMCID: PMC7522597 DOI: 10.3389/fgene.2020.00957] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Recent genome-wide studies of rare genetic variants have begun to implicate novel mechanisms for tetralogy of Fallot (TOF), a severe congenital heart defect (CHD). To provide statistical support for case-only data without parental genomes, we re-analyzed genome sequences of 231 individuals with TOF (n = 175) or related CHD. We adapted a burden test originally developed for de novo variants to assess ultra-rare variant burden in individual genes, and in gene-sets corresponding to functional pathways and mouse phenotypes, accounting for highly correlated gene-sets and for multiple testing. For truncating variants, the gene burden test confirmed significant burden in FLT4 (Bonferroni corrected p-value < 0.01). For missense variants, burden in NOTCH1 achieved genome-wide significance only when restricted to constrained genes (i.e., under negative selection, Bonferroni corrected p-value = 0.004), and showed enrichment for variants affecting the extracellular domain, especially those disrupting cysteine residues forming disulfide bonds (OR = 39.8 vs. gnomAD). Individuals with NOTCH1 ultra-rare missense variants, all with TOF, were enriched for positive family history of CHD. Other genes not previously implicated in CHD had more modest statistical support in gene burden tests. Gene-set burden tests for truncating variants identified a cluster of pathways corresponding to VEGF signaling (FDR = 0%), and of mouse phenotypes corresponding to abnormal vasculature (FDR = 0.8%); these suggested additional candidate genes not previously identified (e.g., WNT5A and ZFAND5). Results for the most promising genes were driven by the TOF subset of the cohort. The findings support the importance of ultra-rare variants disrupting genes involved in VEGF and NOTCH signaling in the genetic architecture of TOF, accounting for 11–14% of individuals in the TOF cohort. These proof-of-principle data indicate that this statistical methodology could assist in analyzing case-only sequencing data in which ultra-rare variants, whether de novo or inherited, contribute to the genetic etiopathogenesis of a complex disorder.
Collapse
Affiliation(s)
- Roozbeh Manshaei
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada
| | - Daniele Merico
- Deep Genomics Inc., Toronto, ON, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Miriam S Reuter
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Worrawat Engchuan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bahareh A Mojarad
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rajiv Chaturvedi
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada.,Labatt Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tracy Heung
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.,The Dalglish Family 22q Clinic, University Health Network, Toronto, ON, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Reem Khan
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada
| | - John B A Okello
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada
| | - Eriskay Liston
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada
| | - Meredith Curtis
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ryan K C Yuen
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada.,Genome Diagnostics, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rebekah K Jobling
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada.,Genome Diagnostics, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Erwin Oechslin
- Division of Cardiology, Toronto Congenital Cardiac Centre for Adults at the Peter Munk Cardiac Centre, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Rachel M Wald
- Labatt Heart Centre, Division of Cardiology, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Cardiology, Toronto Congenital Cardiac Centre for Adults at the Peter Munk Cardiac Centre, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Candice K Silversides
- Division of Cardiology, Toronto Congenital Cardiac Centre for Adults at the Peter Munk Cardiac Centre, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Raymond H Kim
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada.,Fred A. Litwin Family Centre in Genetic Medicine, University Health Network, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.,The Dalglish Family 22q Clinic, University Health Network, Toronto, ON, Canada.,Division of Cardiology, Toronto Congenital Cardiac Centre for Adults at the Peter Munk Cardiac Centre, Department of Medicine, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Mental Health, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
57
|
Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, Carnero A. Therapeutic Targeting of Signaling Pathways Related to Cancer Stemness. Front Oncol 2020; 10:1533. [PMID: 32984007 PMCID: PMC7479251 DOI: 10.3389/fonc.2020.01533] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
The theory of cancer stem cells (CSCs) proposes that the different cells within a tumor, as well as metastasis deriving from it, are originated from a single subpopulation of cells with self-renewal and differentiation capacities. These cancer stem cells are supposed to be critical for tumor expansion and metastasis, tumor relapse and resistance to conventional therapies, such as chemo- and radiotherapy. The acquisition of these abilities has been attributed to the activation of alternative pathways, for instance, WNT, NOTCH, SHH, PI3K, Hippo, or NF-κB pathways, that regulate detoxification mechanisms; increase the metabolic rate; induce resistance to apoptotic, autophagic, and senescence pathways; promote the overexpression of drug transporter proteins; and activate specific stem cell transcription factors. The elimination of CSCs is an important goal in cancer therapeutic approaches because it could decrease relapses and metastatic dissemination, which are main causes of mortality in oncology patients. In this work, we discuss the role of these signaling pathways in CSCs along with their therapeutic potential.
Collapse
Affiliation(s)
- Asunción Espinosa-Sánchez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Elisa Suárez-Martínez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Laura Sánchez-Díaz
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| |
Collapse
|
58
|
Bhopatkar AA, Uversky VN, Rangachari V. Disorder and cysteines in proteins: A design for orchestration of conformational see-saw and modulatory functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 174:331-373. [PMID: 32828470 DOI: 10.1016/bs.pmbts.2020.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Being responsible for more than 90% of cellular functions, protein molecules are workhorses in all the life forms. In order to cater for such a high demand, proteins have evolved to adopt diverse structures that allow them to perform myriad of functions. Beginning with the genetically directed amino acid sequence, the classical understanding of protein function involves adoption of hierarchically complex yet ordered structures. However, advances made over the last two decades have revealed that inasmuch as 50% of eukaryotic proteome exists as partially or fully disordered structures. Significance of such intrinsically disordered proteins (IDPs) is further realized from their ability to exhibit multifunctionality, a feature attributable to their conformational plasticity. Among the coded amino acids, cysteines are considered to be "order-promoting" due to their ability to form inter- or intramolecular disulfide bonds, which confer robust thermal stability to the protein structure in oxidizing conditions. The co-existence of order-promoting cysteines with disorder-promoting sequences seems counter-intuitive yet many proteins have evolved to contain such sequences. In this chapter, we review some of the known cysteine-containing protein domains categorized based on the number of cysteines they possess. We show that many protein domains contain disordered sequences interspersed with cysteines. We show that a positive correlation exists between the degree of cysteines and disorder within the sequences that flank them. Furthermore, based on the computational platform, IUPred2A, we show that cysteine-rich sequences display significant disorder in the reduced but not the oxidized form, increasing the potential for such sequences to function in a redox-sensitive manner. Overall, this chapter provides insights into an exquisite evolutionary design wherein disordered sequences with interspersed cysteines enable potential modulatory protein functions under stress and environmental conditions, which thus far remained largely inconspicuous.
Collapse
Affiliation(s)
- Anukool A Bhopatkar
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, Russia
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS, United States; Center of Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS, United States.
| |
Collapse
|
59
|
Moore G, Annett S, McClements L, Robson T. Top Notch Targeting Strategies in Cancer: A Detailed Overview of Recent Insights and Current Perspectives. Cells 2020; 9:cells9061503. [PMID: 32575680 PMCID: PMC7349363 DOI: 10.3390/cells9061503] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
Evolutionarily conserved Notch plays a critical role in embryonic development and cellular self-renewal. It has both tumour suppressor and oncogenic activity, the latter of which is widely described. Notch-activating mutations are associated with haematological malignancies and several solid tumours including breast, lung and adenoid cystic carcinoma. Moreover, upregulation of Notch receptors and ligands and aberrant Notch signalling is frequently observed in cancer. It is involved in cancer hallmarks including proliferation, survival, migration, angiogenesis, cancer stem cell renewal, metastasis and drug resistance. It is a key component of cell-to-cell interactions between cancer cells and cells of the tumour microenvironment, such as endothelial cells, immune cells and fibroblasts. Notch displays diverse crosstalk with many other oncogenic signalling pathways, and may drive acquired resistance to targeted therapies as well as resistance to standard chemo/radiation therapy. The past 10 years have seen the emergence of different classes of drugs therapeutically targeting Notch including receptor/ligand antibodies, gamma secretase inhibitors (GSI) and most recently, the development of Notch transcription complex inhibitors. It is an exciting time for Notch research with over 70 cancer clinical trials registered and the first-ever Phase III trial of a Notch GSI, nirogacestat, currently at the recruitment stage.
Collapse
Affiliation(s)
- Gillian Moore
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
| | - Stephanie Annett
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
| | - Lana McClements
- The School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
- Correspondence:
| |
Collapse
|
60
|
Xu K, Zhang L. Inhibition of TUG1/miRNA-299-3p Axis Represses Pancreatic Cancer Malignant Progression via Suppression of the Notch1 Pathway. Dig Dis Sci 2020; 65:1748-1760. [PMID: 31655908 DOI: 10.1007/s10620-019-05911-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Taurine-upregulated gene 1 (TUG1) is reported to be upregulated and contributes to the progression of Pancreatic cancer (PC) by serving as an oncogene. Our aims were to explore the precise mechanism of TUG1 involved in PC pathogenesis. METHODS TUG1 and miR-299-3p expression profiles were measured by qRT-PCR. The direct interaction between TUG1 and miR-299-3p was explored by luciferase reporter assay. MTT assay, flow cytometry analysis, caspase-3 activity assay, Transwell invasion assay and wound healing assay were performed to evaluate cell proliferative ability, apoptosis, caspase-3 activity, invasion and migration, respectively. Western blot was conducted to examine the expressions of Ki67, Bax, Bcl-2, matrix metalloproteinase-2 (MMP-2), MMP-9, E-cadherin, N-cadherin, Snail, Notch1, Survivin, and CyclinD1. In addition, animal experiments were also implemented. RESULTS TUG1 was highly expressed, while miR-299-3p was underexpressed in PC tissues and PC cells. Furthermore, the significant increase of TUG1 in PC tissues of advanced patients (stage 3/4) was observed compared to patients (stage 1/2). TUG1 was negatively correlated with miR-299-3p expression in PC tissues. Moreover, TUG1 functioned as a molecular sponge of miR-299-3p to repress its expression. TUG1 knockdown suppressed cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT), and induced apoptosis in PC cells, and repressed tumor growth and EMT in PC xenograft models, which were reversed following reintroduction with anti-miR-299-3p. Furthermore, we found that TUG1 silencing inactivated the Notch1 pathway in PC by upregulating miR-299-3p. CONCLUSIONS The results reported that inhibition of TUG1/miR-299-3p axis suppressed PC malignant progression via suppression of the Notch1 pathway.
Collapse
Affiliation(s)
- Ke Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Νo. 1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lianfeng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Νo. 1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
61
|
Combined inhibition of Notch and FLT3 produces synergistic cytotoxic effects in FLT3/ITD + acute myeloid leukemia. Signal Transduct Target Ther 2020; 5:21. [PMID: 32296014 PMCID: PMC7067872 DOI: 10.1038/s41392-020-0108-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/10/2019] [Accepted: 12/08/2019] [Indexed: 12/30/2022] Open
Abstract
Internal tandem duplication (ITD) mutations of FMS-like tyrosine kinase-3 (FLT3) are the most frequent genetic alterations in acute myeloid leukemia (AML) and predict a poor prognosis. FLT3 tyrosine kinase inhibitors (TKIs) provide short-term clinical responses, but the long-term prognosis of FLT3/ITD+ AML patients remains poor. Notch signaling is important in numerous types of tumors. However, the role of Notch signaling in FLT3/ITD+ AML remains to be elucidated. In the current study, we found that Notch signaling was activated upon FLT3-TKI treatment in FLT3/ITD+ cell lines and primary cells. As Notch signaling can be blocked by γ-secretase inhibitors (GSIs), we examined the combinatorial antitumor efficacy of FLT3-TKIs and GSIs against FLT3/ITD+ AML and explored the underlying molecular mechanisms. As a result, we observed synergistic cytotoxic effects, and the treatment preferentially reduced cell proliferation and induced apoptosis in FLT3/ITD+ AML cell lines and in primary AML cells. Furthermore, the combination of FLT3-TKI and GSI eradicated leukemic cells and prolonged survival in an FLT3/ITD+ patient-derived xenograft AML model. Mechanistically, differential expression analysis suggested that CXCR3 may be partially responsible for the observed synergy, possibly through ERK signaling. Our findings suggest that combined therapies of FLT3-TKIs with GSI may be exploited as a potential therapeutic strategy to treat FLT3/ITD+ AML.
Collapse
|
62
|
Zhang X, Murray B, Mo G, Shern JF. The Role of Polycomb Repressive Complex in Malignant Peripheral Nerve Sheath Tumor. Genes (Basel) 2020; 11:genes11030287. [PMID: 32182803 PMCID: PMC7140867 DOI: 10.3390/genes11030287] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas that can arise most frequently in patients with neurofibromatosis type 1 (NF1). Despite an increasing understanding of the molecular mechanisms that underlie these tumors, there remains limited therapeutic options for this aggressive disease. One potentially critical finding is that a significant proportion of MPNSTs exhibit recurrent mutations in the genes EED or SUZ12, which are key components of the polycomb repressive complex 2 (PRC2). Tumors harboring these genetic lesions lose the marker of transcriptional repression, trimethylation of lysine residue 27 on histone H3 (H3K27me3) and have dysregulated oncogenic signaling. Given the recurrence of PRC2 alterations, intensive research efforts are now underway with a focus on detailing the epigenetic and transcriptomic consequences of PRC2 loss as well as development of novel therapeutic strategies for targeting these lesions. In this review article, we will summarize the recent findings of PRC2 in MPNST tumorigenesis, including highlighting the functions of PRC2 in normal Schwann cell development and nerve injury repair, as well as provide commentary on the potential therapeutic vulnerabilities of a PRC2 deficient tumor cell.
Collapse
Affiliation(s)
- Xiyuan Zhang
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
| | - Béga Murray
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn road, Belfast BT9 7AE, UK
| | - George Mo
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jack F. Shern
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- Correspondence:
| |
Collapse
|
63
|
Mangolini M, Ringshausen I. Bone Marrow Stromal Cells Drive Key Hallmarks of B Cell Malignancies. Int J Mol Sci 2020; 21:E1466. [PMID: 32098106 PMCID: PMC7073037 DOI: 10.3390/ijms21041466] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/06/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
All B cell leukaemias and a substantial fraction of lymphomas display a natural niche residency in the bone marrow. While the bone marrow compartment may only be one of several sites of disease manifestations, the strong clinical significance of minimal residual disease (MRD) in the bone marrow strongly suggests that privileged niches exist in this anatomical site favouring central elements of malignant transformation. Here, the co-existence of two hierarchical systems, originating from haematopoietic and mesenchymal stem cells, has extensively been characterised with regard to regulation of the former (blood production) by the latter. How these two systems cooperate under pathological conditions is far less understood and is the focus of many current investigations. More recent single-cell sequencing techniques have now identified an unappreciated cellular heterogeneity of the bone marrow microenvironment. How each of these cell subtypes interact with each other and regulate normal and malignant haematopoiesis remains to be investigated. Here we review the evidences of how bone marrow stroma cells and malignant B cells reciprocally interact. Evidently from published data, these cell-cell interactions induce profound changes in signalling, gene expression and metabolic adaptations. While the past research has largely focussed on understanding changes imposed by stroma- on tumour cells, it is now clear that tumour-cell contact also has fundamental ramifications for the biology of stroma cells. Their careful characterisations are not only interesting from a scientific biological viewpoint but also relevant to clinical practice: Since tumour cells heavily depend on stroma cells for cell survival, proliferation and dissemination, interference with bone marrow stroma-tumour interactions bear therapeutic potential. The molecular characterisation of tumour-stroma interactions can identify new vulnerabilities, which could be therapeutically exploited.
Collapse
Affiliation(s)
- Maurizio Mangolini
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University hospital, Cambridge CB2 0AH, UK
| |
Collapse
|
64
|
Saini N, Sarin A. Nucleolar localization of the Notch4 intracellular domain underpins its regulation of the cellular response to genotoxic stressors. Cell Death Discov 2020; 6:7. [PMID: 32123583 PMCID: PMC7029026 DOI: 10.1038/s41420-020-0242-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Cell survival is one of the many cellular processes regulated by Notch family of proteins. A comparison of human breast cancer cell lines, which differ in the levels of endogenous Notch4, implicated the protein in regulating susceptibility to apoptosis triggered by genomic damage. In agreement with this observation, increased susceptibility to genotoxic damage was observed following siRNA ablations of Notch4 in two breast cancer cell lines. Further, overexpressing Notch4 intracellular domain (NIC4) tagged to GFP (NIC4-GFP), protected cells from apoptosis triggered by genotoxic drugs. In cells immune-stained for endogenous Notch4, protein was detected in the nucleolus and nucleoplasm, which was also confirmed by the co-localization of NIC4-GFP with RFP-tagged nucleolar proteins in breast cancer cells or the unrelated HEK cell line. Linking functional outcomes to nucleolar localization, NIC4-GFP protection from apoptosis, required the nucleolar proteins Nucleolin and Fibrillarin. Consistently, immunoprecipitation analysis revealed associations between nucleolar proteins-Nucleolin and Nucleophosmin-and Notch4. Microscopy-based biophysical analysis of live cells showed that nucleolar and nucleoplasmic pools of NIC4-GFP are mobile, with some sequestration of nucleolar NIC4-GFP pools. A nucleolar excluded form, NIC4_3RA-GFP, generated by site-directed mutagenesis of the nucleolar localization sequence in NIC4, could not protect from apoptosis triggered by genotoxic stressors. However, transcriptional activity or protection from apoptosis triggered by endoplasmic stress was comparable in cells expressing NIC4_3RA-GFP or NIC4-GFP. Together, the data show that nucleolar localization of NIC4 is critical for the regulation of genomic damage and may be uncoupled from its activities in the nucleoplasm. This study identifies intrinsic features of NIC4 that regulate signaling outcomes activated by the receptor by controlling its spatial localization.
Collapse
Affiliation(s)
- Neetu Saini
- Institute for Stem Cell Science & Regenerative Medicine (inStem), Bellary Road, Bengaluru, Karnataka India
- Department of Biology, Manipal Academy of Higher Education, Manipal, India
| | - Apurva Sarin
- Institute for Stem Cell Science & Regenerative Medicine (inStem), Bellary Road, Bengaluru, Karnataka India
| |
Collapse
|
65
|
Niu K, Guo C, Teng S, Zhou D, Yu S, Yin W, Wang P, Zhu W, Duan M. Pepsin promotes laryngopharyngeal neoplasia by modulating signaling pathways to induce cell proliferation. PLoS One 2020; 15:e0227408. [PMID: 31940393 PMCID: PMC6961942 DOI: 10.1371/journal.pone.0227408] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 12/18/2019] [Indexed: 01/12/2023] Open
Abstract
Pepsin plays an important role in laryngopharyngeal reflux (LPR), a risk factor for the development of hypopharyngeal squamous cell carcinomas (HPSCC). However, the role of pepsin in HPSCC is not clear. We show by immunohistochemistry that pepsin positivity occurs in a significant proportion of human primary HPSCC specimens, and in many cases matched adjacent uninvolved epithelia are negative for pepsin. Pepsin positivity is associated with nodal involvement, suggesting that pepsin may have a role in metastasis. Treatment of FaDu cancer cells with pepsin increased cell proliferation, possibly by inducing G1/S transition. We also observed significant changes in expression of genes involved in NF-kappaB, TRAIL and Notch signaling. Our data suggest that pepsin plays an important role in HPSCC and that targeting pepsin could have potential therapeutic benefits.
Collapse
Affiliation(s)
- Kai Niu
- Department of Otolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, PR China
| | - Chunjie Guo
- Department of Radiology, the First Hospital of Jilin University, Changchun, PR China
| | - Shiyong Teng
- Department of Anesthesiology, the First Hospital of Jilin University, Changchun, PR China
| | - Dandan Zhou
- Department of Radiology, the First Hospital of Jilin University, Changchun, PR China
| | - Shuyuan Yu
- Department of Otolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, PR China
| | - Wanzhong Yin
- Department of Otolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, PR China
| | - Ping Wang
- Department of Otolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, PR China
| | - Wei Zhu
- Department of Otolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, PR China
| | - Maoli Duan
- Department of Clinical Science, Intervention and Technology, Department of Otolaryngology Head and Neck Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
66
|
Xie J, Lin LS, Huang XY, Gan RH, Ding LC, Su BH, Zhao Y, Lu YG, Zheng DL. The NOTCH1-HEY1 pathway regulates self-renewal and epithelial-mesenchymal transition of salivary adenoid cystic carcinoma cells. Int J Biol Sci 2020; 16:598-610. [PMID: 32025208 PMCID: PMC6990919 DOI: 10.7150/ijbs.36407] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Our previous study demonstrated a close relationship between the NOTCH signaling pathway and salivary adenoid cystic carcinoma (SACC). Its receptor gene, NOTCH1, and its downstream gene, HES1, contribute to the proliferation, invasion and metastasis of SACC. Accumulating evidence supports HEY1 as another effector of the signaling pathway. The purpose of this study was to explore the effects of the NOTCH1-HEY1 pathway on the proliferation, invasion and metastasis of SACC cells. Our results verified that HEY1 is a specific molecular target of the NOTCH signaling pathway in SACC cells and that its expression in carcinoma is much higher than that in paracarcinoma tissues. The expression of NOTCH1 and HEY1 are positively correlated in the salivary adenoid cystic carcinoma tissues. NOTCH1 is significantly related to the activation of HEY1 in SACC, and that HEY1 reciprocally regulates NOTCH1 expression in SACC. HEY1 promotes cell proliferation and spheroid formation and inhibits cell apoptosis in vitro. In addition, HEY1 enhances the tumorigenicity of SACC in vivo. Furthermore, HEY1 increases cell invasion and metastasis by driving the expression of epithelial-mesenchymal transition (EMT)-related genes and MMPs. The results of this study indicate that the NOTCH1-HEY1 pathway is specifically upregulated in SACC and promotes cell proliferation, self-renewal, invasion, metastasis and the expression of EMT-related genes and MMPs. Our findings suggest that a NOTCH1-HEY1 pathway inhibitor might therefore have potential therapeutic applications in treating SACC patients by inhibiting cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Jing Xie
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou 350122, China
| | - Li-Song Lin
- Department of Oral and Maxillofacial Surgery, Affiliated First Hospital of Fujian Medical University, 20 Cha Zhong Road, Fuzhou 350005, China
| | - Xiao-Yu Huang
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou 350122, China
| | - Rui-Huan Gan
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| | - Lin-Can Ding
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - Bo-Hua Su
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - Yong Zhao
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China.,Department of pathology, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China
| | - You-Guang Lu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| | - Da-Li Zheng
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou 350004, China
| |
Collapse
|
67
|
Insulin-dependent Non-canonical Activation of Notch in Drosophila: A Story of Notch-Induced Muscle Stem Cell Proliferation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:131-144. [PMID: 32072503 DOI: 10.1007/978-3-030-36422-9_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Notch plays multiple roles both in development and in adult tissue homeostasis. Notch was first identified in Drosophila in which it has then been extensively studied. Among the flag-ship Notch functions we could mention its capacity to keep precursor and stem cells in a nondifferentiated state but also its ability to activate cell proliferation that in some contexts could led to cancer. In general, both these functions involve, canonical, ligand-dependent Notch activation. However, a ligand-independent Notch activation has also been described in a few cellular contexts. Here, we focus on one of such contexts, Drosophila muscle stem cells, called AMPs, and discuss how insulin-dependent noncanonical activation of Notch pushes quiescent AMPs to proliferation.
Collapse
|
68
|
Urata Y, Takeuchi H. Effects of Notch glycosylation on health and diseases. Dev Growth Differ 2019; 62:35-48. [PMID: 31886522 DOI: 10.1111/dgd.12643] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Notch signaling is an evolutionarily conserved signaling pathway and is essential for cell-fate specification in metazoans. Dysregulation of Notch signaling results in various human diseases, including cardiovascular defects and cancer. In 2000, Fringe, a known regulator of Notch signaling, was discovered as a Notch-modifying glycosyltransferase. Since then, glycosylation-a post-translational modification involving literal sugars-on the Notch extracellular domain has been noted as a critical mechanism for the regulation of Notch signaling. Additionally, the presence of diverse O-glycans decorating Notch receptors has been revealed in the extracellular domain epidermal growth factor-like (EGF) repeats. Here, we concisely summarize the recent studies in the human diseases associated with aberrant Notch glycosylation.
Collapse
Affiliation(s)
- Yusuke Urata
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
69
|
Rudan Njavro J, Klotz J, Dislich B, Wanngren J, Shmueli MD, Herber J, Kuhn PH, Kumar R, Koeglsperger T, Conrad M, Wurst W, Feederle R, Vlachos A, Michalakis S, Jedlicka P, Müller SA, Lichtenthaler SF. Mouse brain proteomics establishes MDGA1 and CACHD1 as in vivo substrates of the Alzheimer protease BACE1. FASEB J 2019; 34:2465-2482. [PMID: 31908000 DOI: 10.1096/fj.201902347r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 01/18/2023]
Abstract
The protease beta-site APP cleaving enzyme 1 (BACE1) has fundamental functions in the nervous system. Its inhibition is a major therapeutic approach in Alzheimer's disease, because BACE1 cleaves the amyloid precursor protein (APP), thereby catalyzing the first step in the generation of the pathogenic amyloid beta (Aβ) peptide. Yet, BACE1 cleaves numerous additional membrane proteins besides APP. Most of these substrates have been identified in vitro, but only few were further validated or characterized in vivo. To identify BACE1 substrates with in vivo relevance, we used isotope label-based quantitative proteomics of wild type and BACE1-deficient (BACE1 KO) mouse brains. This approach identified known BACE1 substrates, including Close homolog of L1 and contactin-2, which were found to be enriched in the membrane fraction of BACE1 KO brains. VWFA and cache domain-containing protein 1 (CACHD)1 and MAM domain-containing glycosylphosphatidylinositol anchor protein 1 (MDGA1), which have functions in synaptic transmission, were identified and validated as new BACE1 substrates in vivo by immunoblots using primary neurons and mouse brains. Inhibition or deletion of BACE1 from primary neurons resulted in a pronounced inhibition of substrate cleavage and a concomitant increase in full-length protein levels of CACHD1 and MDGA1. The BACE1 cleavage site in both proteins was determined to be located within the juxtamembrane domain. In summary, this study identifies and validates CACHD1 and MDGA1 as novel in vivo substrates for BACE1, suggesting that cleavage of both proteins may contribute to the numerous functions of BACE1 in the nervous system.
Collapse
Affiliation(s)
- Jasenka Rudan Njavro
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Klotz
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bastian Dislich
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Pathology, University of Bern, Switzerland
| | - Johanna Wanngren
- Division of Neurogeriatrics, Department of NVS, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Merav D Shmueli
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Julia Herber
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Rohit Kumar
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Thomas Koeglsperger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Genome Engineering, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Developmental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Regina Feederle
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Research Center for Environmental Health, Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Zentrum München, Neuherberg, Germany.,Core Facility Monoclonal Antibodies, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Germany.,Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter Jedlicka
- Faculty of Medicine, ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Justus-Liebig-University, Giessen, Germany.,Neuroscience Center, Institute of Clinical Neuroanatomy, Goethe University, Frankfurt am Main, Germany.,Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
70
|
Novel JAG1 Deletion Variant in Patient with Atypical Alagille Syndrome. Int J Mol Sci 2019; 20:ijms20246247. [PMID: 31835735 PMCID: PMC6940840 DOI: 10.3390/ijms20246247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/25/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022] Open
Abstract
Alagille syndrome (AGS) is an autosomal-dominant disorder characterized by various degrees of abnormalities in the liver, heart, eyes, vertebrae, kidneys, face, vasculature, skeleton, and pancreas. This case report describes a newborn child exhibiting a congenital neural tube defect and peculiar craniofacial appearance characterized by a prominent forehead, deep-set eyes, bulbous nasal tip, and subtle upper lip. Just a few hours after birth, congenital heart disease was suspected for cyanosis and confirmed by heart evaluation. In particular, echocardiography indicated pulmonary atresia with ventricular septal defect with severe hypoplasia of the pulmonary branches (1.5 mm), large patent ductus arteriosus and several major aortopulmonary collateral arteries. Due to the association of peculiar craniofacial appearance and congenital heart disease, a form of Alagille syndrome was suspected. In addition, on the fifth day after birth, the patient developed jaundice, had acholic stools, and high levels of conjugated bilirubin and gamma-glutamyltransferase (GGT) were detected in the blood. Genetic testing revealed the novel variant c.802del in a single copy of the JAG1 gene. No variants in the NOTCH2 gene were detected. To the best of our knowledge, this is the first clinical description of a congenital neural tube defect in a molecularly confirmed Alagille patient. This work demonstrates a novel pathogenic heterozygous JAG1 mutation is associated with an atypical form of Alagille syndrome, suggesting an increased risk for neural tube defects compared to other Alagille patients.
Collapse
|
71
|
Das P, Salazar JL, Li-Kroeger D, Yamamoto S, Nakamura M, Sasamura T, Inaki M, Masuda W, Kitagawa M, Yamakawa T, Matsuno K. Maternal almondex, a neurogenic gene, is required for proper subcellular Notch distribution in early Drosophila embryogenesis. Dev Growth Differ 2019; 62:80-93. [PMID: 31782145 DOI: 10.1111/dgd.12639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/08/2019] [Indexed: 01/03/2023]
Abstract
Notch signaling plays crucial roles in the control of cell fate and physiology through local cell-cell interactions. The core processes of Notch signal transduction are well established, but the mechanisms that fine-tune the pathway in various developmental and post-developmental contexts are less clear. Drosophila almondex, which encodes an evolutionarily conserved double-pass transmembrane protein, was identified in the 1970s as a maternal-effect gene that regulates Notch signaling in certain contexts, but its mechanistic function remains obscure. In this study, we examined the role of almondex in Notch signaling during early Drosophila embryogenesis. We found that in addition to being required for lateral inhibition in the neuroectoderm, almondex is also partially required for Notch signaling-dependent single-minded expression in the mesectoderm. Furthermore, we found that almondex is required for proper subcellular Notch receptor distribution in the neuroectoderm, specifically during mid-stage 5 development. The absence of maternal almondex during this critical window of time caused Notch to accumulate abnormally in cells in a mesh-like pattern. This phenotype did not include any obvious change in subcellular Delta ligand distribution, suggesting that it does not result from a general vesicular-trafficking defect. Considering that dynamic Notch trafficking regulates signal output to fit the specific context, we speculate that almondex may facilitate Notch activation by regulating intracellular Notch receptor distribution during early embryogenesis.
Collapse
Affiliation(s)
- Puspa Das
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Jose L Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - David Li-Kroeger
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Mitsutoshi Nakamura
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Takeshi Sasamura
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Mikiko Inaki
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Wataru Masuda
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Motoo Kitagawa
- Department of Biochemistry, International University of Health and Welfare, School of Medicine, Chiba, Japan
| | - Tomoko Yamakawa
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Kenji Matsuno
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| |
Collapse
|
72
|
Saad MI, Rose-John S, Jenkins BJ. ADAM17: An Emerging Therapeutic Target for Lung Cancer. Cancers (Basel) 2019; 11:E1218. [PMID: 31438559 PMCID: PMC6769596 DOI: 10.3390/cancers11091218] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 12/23/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality, which histologically is classified into small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC accounts for approximately 85% of all lung cancer diagnoses, with the majority of patients presenting with lung adenocarcinoma (LAC). KRAS mutations are a major driver of LAC, and are closely related to cigarette smoking, unlike mutations in the epidermal growth factor receptor (EGFR) which arise in never-smokers. Although the past two decades have seen fundamental progress in the treatment and diagnosis of NSCLC, NSCLC still is predominantly diagnosed at an advanced stage when therapeutic interventions are mostly palliative. A disintegrin and metalloproteinase 17 (ADAM17), also known as tumour necrosis factor-α (TNFα)-converting enzyme (TACE), is responsible for the protease-driven shedding of more than 70 membrane-tethered cytokines, growth factors and cell surface receptors. Among these, the soluble interleukin-6 receptor (sIL-6R), which drives pro-inflammatory and pro-tumourigenic IL-6 trans-signaling, along with several EGFR family ligands, are the best characterised. This large repertoire of substrates processed by ADAM17 places it as a pivotal orchestrator of a myriad of physiological and pathological processes associated with the initiation and/or progression of cancer, such as cell proliferation, survival, regeneration, differentiation and inflammation. In this review, we discuss recent research implicating ADAM17 as a key player in the development of LAC, and highlight the potential of ADAM17 inhibition as a promising therapeutic strategy to tackle this deadly malignancy.
Collapse
Affiliation(s)
- Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, D-24098 Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
73
|
Papadakos KS, Bartoschek M, Rodriguez C, Gialeli C, Jin SB, Lendahl U, Pietras K, Blom AM. Cartilage Oligomeric Matrix Protein initiates cancer stem cells through activation of Jagged1-Notch3 signaling. Matrix Biol 2019; 81:107-121. [DOI: 10.1016/j.matbio.2018.11.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/30/2022]
|
74
|
Qin S, Zheng JH, Xia ZH, Qian J, Deng CL, Yang SL. CTHRC1 promotes wound repair by increasing M2 macrophages via regulating the TGF-β and notch pathways. Biomed Pharmacother 2019; 113:108594. [PMID: 30849639 DOI: 10.1016/j.biopha.2019.01.055] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
The healing of acute wounds is vital to humans and is a well-orchestrated process that involves systemic and local factors. However, there is a lack of effective and safe clinical therapies. The collagen triple helix repeat containing 1 (CTHRC1) protein is a type of exocrine protein that has been recently reported to contribute to tissue repair. Our aim is to validate the promoting effects of CTHRC1 on the healing of acute wounds and to elucidate the underlying molecular mechanism. Therefore, we first established acute wound healing mouse models and confirmed that CTHRC1 accelerates the healing process of acute wounds. Then, we characterized wound macrophages using a polyvinylalcohol (PVA) sponge model and used Western blotting to investigate the molecular mechanism. We found that CTHRC1 increased the M2 macrophage population and the TGF-β expression level as a result of the activation of the TGF-β and Notch pathways, which eventually contributed to the promotion of wound healing. Inhibition of the Notch pathway showed attenuated M2 macrophage recruitment, and it decreased the TGF-β expression level. These results substantiate our hypothesis that CTHRC1 promotes wound healing by recruiting M2 macrophages and regulating the TGF-β and Notch pathways.
Collapse
Affiliation(s)
- Shu Qin
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Jiang-Hong Zheng
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Zi-Huan Xia
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Jin Qian
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Chen-Liang Deng
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Song-Lin Yang
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| |
Collapse
|
75
|
Henrique D, Schweisguth F. Mechanisms of Notch signaling: a simple logic deployed in time and space. Development 2019; 146:146/3/dev172148. [PMID: 30709911 DOI: 10.1242/dev.172148] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Most cells in our body communicate during development and throughout life via Notch receptors and their ligands. Notch receptors relay information from the cell surface to the genome via a very simple mechanism, yet Notch plays multiple roles in development and disease. Recent studies suggest that this versatility in Notch function may not necessarily arise from complex and context-dependent integration of Notch signaling with other developmental signals, but instead arises, in part, from signaling dynamics. Here, we review recent findings on the core Notch signaling mechanism and discuss how spatial-temporal dynamics contribute to Notch signaling output.
Collapse
Affiliation(s)
- Domingos Henrique
- Instituto de Histologia e Biologia do Desenvolvimento and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egaz Moniz, 1649-028 Lisboa, Portugal
| | - François Schweisguth
- Institut Pasteur, Department of Developmental and Stem Cell Biology, F-75015 Paris, France .,CNRS, UMR3738, F-75015 Paris, France
| |
Collapse
|
76
|
Maier D. The evolution of transcriptional repressors in the Notch signaling pathway: a computational analysis. Hereditas 2019; 156:5. [PMID: 30679936 PMCID: PMC6337844 DOI: 10.1186/s41065-019-0081-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/09/2019] [Indexed: 11/18/2022] Open
Abstract
Background The Notch signaling pathway governs the specification of different cell types in flies, nematodes and vertebrates alike. Principal components of the pathway that activate Notch target genes are highly conserved throughout the animal kingdom. Despite the impact on development and disease, repression mechanisms are less well studied. Repressors are known from arthropods and vertebrates that differ strikingly by mode of action: whereas Drosophila Hairless assembles repressor complexes with CSL transcription factors, competition between activator and repressors occurs in vertebrates (for example SHARP/MINT and KyoT2). This divergence raises questions on the evolution: Are there common ancestors throughout the animal kingdom? Results Available genome databases representing all animal clades were searched for homologues of Hairless, SHARP and KyoT2. The most distant species with convincing Hairless orthologs belong to Myriapoda, indicating its emergence after the Mandibulata-Chelicarata radiation about 500 million years ago. SHARP shares motifs with SPEN and SPENITO proteins, present throughout the animal kingdom. The CSL interacting domain of SHARP, however, is specific to vertebrates separated by roughly 600 million years of evolution. KyoT2 bears a C-terminal CSL interaction domain (CID), present only in placental mammals but highly diverged already in marsupials, suggesting introduction roughly 100 million years ago. Based on the LIM-domains that characterize KyoT2, homologues can be found in Drosophila melanogaster (Limpet) and Hydra vulgaris (Prickle 3 like). These lack the CID of KyoT2, however, contain a PET and additional LIM domains. Conservation of intron/exon boundaries underscores the phylogenetic relationship between KyoT2, Limpet and Prickle. Most strikingly, Limpet and Prickle proteins carry a tetra-peptide motif resembling that of several CSL interactors. Overall, KyoT2 may have evolved from prickle and Limpet to a Notch repressor in mammals. Conclusions Notch repressors appear to be specific to either chordates or arthropods. Orthologues of experimentally validated repressors were not found outside the phylogenetic group they have been originally identified. However, the data provide a hypothesis on the evolution of mammalian KyoT2 from Prickle like ancestors. The finding of a potential CSL interacting domain in Prickle homologues points to a novel, very ancestral CSL interactor present in the entire animal kingdom. Electronic supplementary material The online version of this article (10.1186/s41065-019-0081-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dieter Maier
- Institute of Genetics (240), University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
77
|
Alabi RO, Farber G, Blobel CP. Intriguing Roles for Endothelial ADAM10/Notch Signaling in the Development of Organ-Specific Vascular Beds. Physiol Rev 2019; 98:2025-2061. [PMID: 30067156 DOI: 10.1152/physrev.00029.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The vasculature is a remarkably interesting, complex, and interconnected organ. It provides a conduit for oxygen and nutrients, filtration of waste products, and rapid communication between organs. Much remains to be learned about the specialized vascular beds that fulfill these diverse, yet vital functions. This review was prompted by the discovery that Notch signaling in mouse endothelial cells is crucial for the development of specialized vascular beds found in the heart, kidneys, liver, intestines, and bone. We will address the intriguing questions raised by the role of Notch signaling and that of its regulator, the metalloprotease ADAM10, in the development of specialized vascular beds. We will cover fundamentals of ADAM10/Notch signaling, the concept of Notch-dependent cell fate decisions, and how these might govern the development of organ-specific vascular beds through angiogenesis or vasculogenesis. We will also consider common features of the affected vessels, including the presence of fenestra or sinusoids and their occurrence in portal systems with two consecutive capillary beds. We hope to stimulate further discussion and study of the role of ADAM10/Notch signaling in the development of specialized vascular structures, which might help uncover new targets for the repair of vascular beds damaged in conditions like coronary artery disease and glomerulonephritis.
Collapse
Affiliation(s)
- Rolake O Alabi
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| | - Gregory Farber
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| | - Carl P Blobel
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| |
Collapse
|
78
|
Zubeldía-Brenner L, De Winne C, Perrone S, Rodríguez-Seguí SA, Willems C, Ornstein AM, Lacau-Mengido I, Vankelecom H, Cristina C, Becu-Villalobos D. Inhibition of Notch signaling attenuates pituitary adenoma growth in Nude mice. Endocr Relat Cancer 2019; 26:13-29. [PMID: 30121620 DOI: 10.1530/erc-18-0337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/08/2018] [Indexed: 12/16/2022]
Abstract
Preclinical and clinical studies support that Notch signaling may play an important oncogenic role in cancer, but there is scarce information for pituitary tumors. We therefore undertook a functional study to evaluate Notch participation in pituitary adenoma growth. Tumors generated in Nude mice by subcutaneous GH3 somatolactotrope cell injection were treated in vivo with DAPT, a γ-secretase inhibitor, thus inactivating Notch signaling. This treatment led to pituitary tumor reduction, lower prolactin and GH tumor content and a decrease in angiogenesis. Furthermore, in silico transcriptomic and epigenomic analyses uncovered several tumor suppressor genes related to Notch signaling in pituitary tissue, namely Btg2, Nr4a1, Men1, Zfp36 and Cnot1. Gene evaluation suggested that Btg2, Nr4a1 and Cnot1 may be possible players in GH3 xenograft growth. Btg2 mRNA expression was lower in GH3 tumors compared to the parental line, and DAPT increased its expression levels in the tumor in parallel with the inhibition of its volume. Cnot1 mRNA levels were also increased in the pituitary xenografts by DAPT treatment. And the Nr4a1 gene was lower in tumors compared to the parental line, though not modified by DAPT. Finally, because DAPT in vivo may also be acting on tumor microenvironment, we determined the direct effect of DAPT on GH3 cells in vitro. We found that DAPT decreases the proliferative, secretory and migration potential of GH3 cells. These results position selective interruption of Notch signaling as a potential therapeutic tool in adjuvant treatments for aggressive or resistant pituitary tumors.
Collapse
Affiliation(s)
| | - Catalina De Winne
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | - Sofía Perrone
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, CITNOBA (UNNOBA-CONICET), Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Santiago A Rodríguez-Seguí
- Departamento de Fisiología y Biología Molecular y Celular, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina
| | - Christophe Willems
- Department of Development and Regeneration, Cluster Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Ana María Ornstein
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | - Isabel Lacau-Mengido
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Carolina Cristina
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, CITNOBA (UNNOBA-CONICET), Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
79
|
Takam Kamga P, Dal Collo G, Midolo M, Adamo A, Delfino P, Mercuri A, Cesaro S, Mimiola E, Bonifacio M, Andreini A, Chilosi M, Krampera M. Inhibition of Notch Signaling Enhances Chemosensitivity in B-cell Precursor Acute Lymphoblastic Leukemia. Cancer Res 2018; 79:639-649. [PMID: 30563887 DOI: 10.1158/0008-5472.can-18-1617] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 10/23/2018] [Accepted: 12/14/2018] [Indexed: 11/16/2022]
Abstract
Notch3 and Notch4 support survival of primary B-cell acute lymphoblastic leukemia (B-ALL) cells, suggesting a role for Notch signaling in drug response. Here we used in vitro, in silico, and in vivo mouse xenograft model-based approaches to define the role of the Notch pathway in B-ALL chemosensitivity. We observed significant Notch receptor and ligand expression in B-ALL primary cells and cell lines. Primary leukemia cells from high-risk patients overexpressed Notch3, Notch4, and Jagged2 while displaying a reduction in expression levels of Notch1-4 following chemotherapy. We then analyzed in vitro cell survival of B-ALL cells treated with conventional chemotherapeutic agents alone or in combination with Notch signaling inhibitors. Gamma-secretase inhibitors (GSI) and anti-Notch4 were all capable of potentiating drug-induced cell death in B-ALL cells by upregulating intracellular levels of reactive oxygen species, which in turn modulated mTOR, NF-κB, and ERK expression. In NOG-mouse-based xenograft models of B-ALL, co-administration of the Notch inhibitor GSI-XII with the chemotherapeutic agent Ara-C lowered bone marrow leukemic burden compared with DMSO or Ara-C alone, thus prolonging mouse survival. Overall, our results support the potential effectiveness of Notch inhibitors in patients with B-ALL.Significance: Inhibition of Notch signaling enhances the chemosensitivity of B-ALL cells, suggesting Notch inhibition as a potential therapeutic strategy to improve the outcome of patients with B-ALL.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.,EA4340-BCOH: Biomarker in Cancerology and Onco-Hematology, Université de Versailles Saint-Quentin-en-Yvelines, Versailles, France
| | - Giada Dal Collo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Martina Midolo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Annalisa Adamo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Pietro Delfino
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Angela Mercuri
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.,Pediatric Onco-Hematology Unit, University of Verona, Verona, Italy
| | - Simone Cesaro
- Pediatric Onco-Hematology Unit, University of Verona, Verona, Italy
| | - Elda Mimiola
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Massimiliano Bonifacio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Angelo Andreini
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Marco Chilosi
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
80
|
Arruga F, Vaisitti T, Deaglio S. The NOTCH Pathway and Its Mutations in Mature B Cell Malignancies. Front Oncol 2018; 8:550. [PMID: 30534535 PMCID: PMC6275466 DOI: 10.3389/fonc.2018.00550] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/06/2018] [Indexed: 12/16/2022] Open
Abstract
The systematic application of next-generation sequencing to large cohorts of oncologic samples has opened a Pandora's box full of known and novel genetic lesions implicated in different steps of cancer development and progression. Narrowing down to B cell malignancies, many previously unrecognized genes emerged as recurrently mutated. The challenge now is to determine how the mutation in a given gene affects the biology of the disease, paving the way to functional genomics studies. Mutations in NOTCH family members are shared by several disorders of the B series, even if with variable frequencies and mutational patterns. In silico predictions, revealed that mutations occurring in NOTCH receptors, despite being qualitatively different, may have similar effects on protein processing, ultimately leading to enhanced pathway activation. The discovery of mutations occurring also in downstream players, either potentiating positive signals or compromising negative regulators, indicates that multiple mechanisms in neoplastic B cells concur to activate NOTCH pathway. These findings are supported by results obtained in chronic lymphocytic leukemia and splenic marginal zone B cell lymphoma where deregulation of NOTCH signaling has been functionally characterized. The emerging picture confirms that NOTCH signaling is finely tuned in cell- and microenvironment-dependent ways. In B cell malignancies, it contributes to the regulation of proliferation, survival and migration. However, deeper biological studies are needed to pinpoint the contribution of NOTCH in the hierarchy of events driving B cells transformation, keeping in mind its role in normal B cells development. Because of its relevance in leukemia and lymphoma biology, the NOTCH pathway might represent an appealing therapeutic target: the next few years will tell whether this potential will be fulfilled.
Collapse
Affiliation(s)
- Francesca Arruga
- Italian Institute for Genomic Medicine, Turin, Italy.,Department of Medical Sciences, University of Torino, Turin, Italy
| | - Tiziana Vaisitti
- Italian Institute for Genomic Medicine, Turin, Italy.,Department of Medical Sciences, University of Torino, Turin, Italy
| | - Silvia Deaglio
- Italian Institute for Genomic Medicine, Turin, Italy.,Department of Medical Sciences, University of Torino, Turin, Italy
| |
Collapse
|
81
|
Gera S, Dighe RR. The soluble ligand Y box-1 activates Notch3 receptor by binding to epidermal growth factor like repeats 20-23. Arch Biochem Biophys 2018; 660:129-136. [PMID: 30321499 DOI: 10.1016/j.abb.2018.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/14/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022]
Abstract
The transduction of signal by the Notch receptors to the intracellular domain is highly regulated and relies on binding of the ligands to the Epidermal growth factor Like Repeats (ELRs) of receptor's extracellular domain. Both canonical and non-canonical ligands are known to interact with different ELRs and activate Notch receptors. The aim of this study was to investigate the interaction of a soluble non-canonical ligand, Y box-1 (Yb-1) with Notch3 receptor ELRs. Polyclonal antibodies were employed as novel tools to identify the binding site of this ligand. Using various ligand binding and signaling assays, soluble Yb-1 was found to interact specifically with the Notch3 receptor, but not with Notch1. The ELRs 17-24 of Notch3 were identified as the binding site for Yb-1. Further, Yb-1 and Notch3 ELRs 17-24 structures were modelled and the Yb-1-Notch3 interaction interface was predicted to be Notch3 ELRs 20-23. Binding of the Yb-1 with Notch3 ELRs different from those reported for canonical DSL ligands also transduced the signal to the intracellular domain through the negative regulatory region. In conclusion, study highlights the importance of molecular modifications in different Notch3 ELRs for the transduction of signal to the negative regulatory region.
Collapse
Affiliation(s)
- Sakshi Gera
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Rajan R Dighe
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
82
|
Zhang Q, Zhou J, Lei H, Zhu CY, Li FF, Zheng D, Liu SL. RBPJ polymorphisms associated with cerebral infarction diseases in Chinese Han population: A Clinical Trial/Experimental Study (CONSORT Compliant). Medicine (Baltimore) 2018; 97:e11420. [PMID: 30075508 PMCID: PMC6081149 DOI: 10.1097/md.0000000000011420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
TRIAL DESIGN Cerebral small vessel diseases (CSVDs) are a group of brain pathological processes involving cerebral small arteries, brain venules, and capillaries. The recombination signal-binding protein Jκ (RBPJ) is implicated in the pathogenesis of these diseases but its actual roles need confirmation. The aim of this work was to evaluate variations in RBPJ gene for their possible associations with the disease. METHODS The RBPJ gene was sequenced for 400 patients with cerebral infarction disease and 600 normal controls. The statistical analyses and Hardy-Weinberg equilibrium tests of the patients and control populations were conducted using the SPSS software (version 19.0) and Plink (version 1.9), Haploview software, and online software SNPSpD. RESULTS We characterized variants rs2871198, rs1397731, rs3822223, rs2077777, rs2270226, and rs2788861 within or near the RBPJ gene. The genetic heterozygosity of rs2871198, rs1397731, rs3822223, rs2077777, and rs2270226 was very high. Statistical analysis showed that the variants rs2270226 and rs2077777 in the gene were associated with the risk of cerebral infarction diseases in the Chinese Han population. CONCLUSIONS rs2270226 and rs2077777 in the RBPJ gene were associated with the risk of cerebral infarction diseases in the Chinese Han population.
Collapse
Affiliation(s)
- Qiong Zhang
- College of Wildlife Resources, Northeast Forestry University
- Department of Antibiotics, Heilongjiang Institute for Food and Drug Control
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
| | - Jie Zhou
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Hong Lei
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Chun-Yu Zhu
- Department of Neurology, Daqing Oilfield General Hospital, Daqing, China
| | - Fei-Feng Li
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Dong Zheng
- College of Wildlife Resources, Northeast Forestry University
| | - Shu-Lin Liu
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
83
|
Gambucci M, Tarpani L, Zampini G, Massaro G, Nocchetti M, Sassi P, Latterini L. Fluorimetric Studies of a Transmembrane Protein and Its Interactions with Differently Functionalized Silver Nanoparticles. J Phys Chem B 2018; 122:6872-6879. [PMID: 29911868 DOI: 10.1021/acs.jpcb.8b02599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transmembrane proteins play important roles in intercellular signaling to regulate interactions among the adjacent cells and influence cell fate. The study of interactions between membrane proteins and nanomaterials is paramount for the design of nanomaterial-based therapies. In the present work, the fluorescence properties of the transmembrane receptor Notch2 have been investigated. In particular, the steady-state and time-resolved fluorescence methods have been used to characterize the emission of tryptophan residues of Notch2 and then this emission is used to monitor the effect of silver colloids on protein behavior. To this aim, silver colloids are prepared with two different methods to make sure that they bear hydrophilic (citrate ions, C-AgNPs) or hydrophobic (dodecanethiol molecules, D-AgNPs) capping agents. The preparation procedures are tightly controlled to obtain metal cores with similar size distributions (7.4 ± 2.5 and 5.0 ± 0.8 nm, respectively), thus, making the comparison of the results easier. The occurrence of strong interactions between Notch2 and D-AgNPs is suggested by the efficient and statistically relevant quenching of the stationary protein emission already at low nanoparticle (NP) concentrations (ca. 12% quenching with [D-AgNPs] = 0.6 nM). The quenching becomes even more pronounced (ca. 60%) when [D-AgNPs] is raised to 8.72 nM. On the other hand, the addition of increasing concentrations of C-AgNPs to Notch2 does not affect the protein fluorescence (intensity variations below 5%) indicating that negligible interactions are taking place. The fluorescence data, recorded in the presence of increasing concentrations of silver nanoparticles, are then analyzed through the Stern-Volmer equation and the sphere of action model to discuss the nature of interactions. The effect of D-AgNPs on the fluorescence decay times of Notch2 is also investigated and a decrease in the average decay time is observed (from 4.64 to 3.42 ns). The observed variations of the stationary and time-resolved fluorescence behavior of the protein are discussed in terms of static and collisional interactions. These results document that the capping shell is able to drive the protein-particle interactions, which likely have a hydrophobic nature.
Collapse
Affiliation(s)
- Marta Gambucci
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| | - Luigi Tarpani
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| | - Giulia Zampini
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| | - Giuseppina Massaro
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| | - Morena Nocchetti
- Dipartimento di Scienze Farmaceutiche , Università di Perugia , Via del Liceo 1 , 06123 Perugia , Italy
| | - Paola Sassi
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| | - Loredana Latterini
- Dipartimento di Chimica, Biologia e Biotecnologie , Università di Perugia , Via Elce di Sotto, 8 , 06123 Perugia , Italy
| |
Collapse
|
84
|
Kamga PT, Dal Collo G, Bassi G, Midolo M, Delledonne M, Chilosi M, Bonifacio M, Krampera M. Characterization of a new B-ALL cell line with constitutional defect of the Notch signaling pathway. Oncotarget 2018; 9:18341-18350. [PMID: 29719609 PMCID: PMC5915076 DOI: 10.18632/oncotarget.24836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/11/2018] [Indexed: 12/31/2022] Open
Abstract
Notch signaling contribution to B-cell acute lymphoblastic leukemia (B-ALL)
development is still under investigation. The serendipitous onset of B-ALL in a
patient affected by the germinal Notch mutation-dependent Alagille syndrome allowed
us to establish a B-ALL cell line (VR-ALL) bearing a genetic loss of function in
components of Notch signaling. VR-ALL is a common-type B-ALL cell line, grows in
conventional culture medium supplemented with 10% serum, and gives rise, once
injected into immunodeficient NOG mice, to a mouse xenograft model of B-ALL. Exome
sequencing revealed deleterious mutations in some components of Notch signaling,
including Jagged1, Notch1, and Notch2. In addition, VR-ALL is sensitive both
in vitro and in vivo to γ-secretase
inhibitors (GSIs) as well as conventional anti-leukemic drugs. For all these reasons,
VR-ALL may help to gain more insights into the role of Notch signaling in B-ALL.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giada Dal Collo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giulio Bassi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Martina Midolo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Massimo Delledonne
- Department of Biotechnology, University of Verona, Verona, Italy.,Personal Genomics S.R.L., Verona, Italy
| | - Marco Chilosi
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Massimiliano Bonifacio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
85
|
Rossini M, Rizzo P, Bononi I, Clementz A, Ferrari R, Martini F, Tognon MG. New Perspectives on Diagnosis and Therapy of Malignant Pleural Mesothelioma. Front Oncol 2018; 8:91. [PMID: 29666782 PMCID: PMC5891579 DOI: 10.3389/fonc.2018.00091] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/15/2018] [Indexed: 12/24/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, but severe form of cancer, with an incidence that varies significantly within and among different countries around the world. It develops in about one to two persons per million of the general population, leading to thousands of deaths every year worldwide. To date, the MPM is mostly associated with occupational asbestos exposure. Asbestos represents the predominant etiological factor, with approximately 70% of cases of MPM with well-documented occupational exposure to asbestos, with the exposure time, on average greater than 40 years. Environmental exposure to asbestos is increasingly becoming recognized as a cause of mesothelioma, together with gene mutations. The possible roles of other cofactors, such as viral infection and radiation exposure, are still debated. MPM is a fatal tumor. This cancer arises during its early phase without clinical signs. Consequently, its diagnosis occurs at advanced stages. Standard clinical therapeutic approaches include surgery, chemo- and radiotherapies. Preclinical and clinical researches are making great strides in the field of this deadly disease, identifying new biomarkers and innovative therapeutic approaches. Among the newly identified markers and potential therapeutic targets, circulating microRNAs and the Notch pathway represent promising avenues that could result in the early detection of the tumor and novel therapeutic approaches.
Collapse
Affiliation(s)
- Marika Rossini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Bononi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Anthony Clementz
- Department of Natural Sciences and Geography, Concordia University Chicago, River Forest, IL, United States
| | - Roberto Ferrari
- Department of Medical Sciences, Section of Internal Medicine and Cardiorespiratory, School of Medicine, University of Ferrara, Ferrara, Italy.,E.S. Health Science Foundation, GVM Care & Research, Maria Cecilia Hospital, Cotignola, Italy
| | - Fernanda Martini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro G Tognon
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
86
|
Abstract
Comprehensive genomic analyses have been performed for head and neck squamous cell carcinoma (HNSCC), revealing a significant rate of NOTCH1 mutations and identifying NOTCH1 as the second most frequently mutated gene after TP53. Most NOTCH1 mutations are considered inactivating, indicating that NOTCH1 is a tumor suppressor gene. On the other hand, cohorts from Asian populations with HNSCC have shown activating NOTCH1 mutations. HNSCC with NOTCH1 mutations have a worse prognosis than the NOTCH1 wild-type tumors. Additional data on other NOTCH family members have shown that NOTCH promotes HNSCC progression. NOTCH family members, including NOTCH pathway genes, are upregulated in HNSCC compared with normal tissues, and inhibition of the NOTCH pathway decreases cell proliferation and invasion. NOTCH activity in HNSCC is therefore contextual, and NOTCH in HNSCC is considered to have a bimodal role as a tumor suppressor and an oncogene. In this review, recent understandings of NOTCH pathway genes, including NOTCH genes, in HNSCC are described. In addition, the implications of NOTCH pathway alteration for HNSCC-specific NOTCH-targeted cancer therapy are explored.
Collapse
Affiliation(s)
- T Fukusumi
- 1 Moores Cancer Center, University of California, La Jolla, CA, USA
| | - J A Califano
- 1 Moores Cancer Center, University of California, La Jolla, CA, USA
| |
Collapse
|
87
|
Notch pathway signaling in the skin antagonizes Merkel cell development. Dev Biol 2018; 434:207-214. [DOI: 10.1016/j.ydbio.2017.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 01/16/2023]
|
88
|
Epsin-Dependent Ligand Endocytosis Activates Notch by Force. Cell 2017; 171:1383-1396.e12. [PMID: 29195077 DOI: 10.1016/j.cell.2017.10.048] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/30/2017] [Accepted: 09/28/2017] [Indexed: 01/09/2023]
Abstract
DSL ligands activate Notch by inducing proteolytic cleavage of the receptor ectodomain, an event that requires ligand to be endocytosed in signal-sending cells by the adaptor protein Epsin. Two classes of explanation for this unusual requirement are (1) recycling models, in which the ligand must be endocytosed to be modified or repositioned before it binds Notch and (2) pulling models, in which the ligand must be endocytosed after it binds Notch to exert force that exposes an otherwise buried site for cleavage. We demonstrate in vivo that ligands that cannot enter the Epsin pathway nevertheless bind Notch but fail to activate the receptor because they cannot exert sufficient force. This argues against recycling models and in favor of pulling models. Our results also suggest that once ligand binds receptor, activation depends on a competition between Epsin-mediated ligand endocytosis, which induces cleavage, and transendocytosis of the ligand by the receptor, which aborts the incipient signal.
Collapse
|
89
|
Gan RH, Wei H, Xie J, Zheng DP, Luo EL, Huang XY, Xie J, Zhao Y, Ding LC, Su BH, Lin LS, Zheng DL, Lu YG. Notch1 regulates tongue cancer cells proliferation, apoptosis and invasion. Cell Cycle 2017; 17:216-224. [PMID: 29117785 DOI: 10.1080/15384101.2017.1395534] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Notch1 regulates tumor biology in a complex, context-dependent manner. The roles of Notch1 in tongue cancer are still controversial. The aim of this study is to investigate the roles of Notch1 in tongue cancer. MATERIALS AND METHODS The expression of Notch1 was tested between tongue cancer and normal samples by using immunohistochemistry. Tongue cancer cells were transfected with siRNA or plasmid, respectively. Cell proliferation, apoptosis, migration and invasion ability were tested in appropriate ways. The subcutaneous tumor model was established to observe the tumor growth. RESULTS Notch1 was upregulated in tongue carcinoma tissues and the expression of Notch1 was related with tumor stage and differentiation. Overexpression of Notch1 could increase tongue cancer cells proliferation, invasion and migration. But inhibited the expression of Notch1 could decrease cells proliferation, invasion and migration and promote cell apoptosis in vitro and in vivo. CONCLUSION Our results prove that the oncogenic role of Notch1 in tongue cancer and provide the direction of targeted therapy of tongue cancer.
Collapse
Affiliation(s)
- Rui-Huan Gan
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Hua Wei
- c The School of Basic Medical Science , Fujian Medical University , 1 Xue Yuan Road, Shang Jie Town, Min Hou County, Fuzhou 350000 , China
| | - Jing Xie
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Dan-Ping Zheng
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Er-Ling Luo
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Xiao-Yu Huang
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Jian Xie
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Yong Zhao
- b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China.,d Department of Pathology , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China
| | - Lin-Can Ding
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Bo-Hua Su
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| | - Li-Song Lin
- e Department of Oral and Maxillofacial Surgery , Affiliated First Hospital of Fujian Medical University , 20 Cha Zhong Road, Fuzhou 350000 , China
| | - Da-Li Zheng
- f Department of Molecular Medicine , University of South Florida , 19202 Bruce B Downs Blvd, Tampa , FL 33613 , United States
| | - You-Guang Lu
- a Department of Preventive Dentistry , Affiliated Stomatological Hospital, Fujian Medical University , 246 Yang Qiao Middle Road, Fuzhou 350000 , China.,b Key laboratory of stomatology , Fujian Province University , 88 Jiao Tong Road, Fuzhou 350004 , China
| |
Collapse
|
90
|
Gazave E, Lemaître QIB, Balavoine G. The Notch pathway in the annelid Platynereis: insights into chaetogenesis and neurogenesis processes. Open Biol 2017; 7:rsob.160242. [PMID: 28148821 PMCID: PMC5356439 DOI: 10.1098/rsob.160242] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023] Open
Abstract
Notch is a key signalling pathway playing multiple and varied functions during development. Notch regulates the selection of cells with a neurogenic fate and maintains a pool of yet uncommitted precursors through lateral inhibition, both in insects and in vertebrates. Here, we explore the functions of Notch in the annelid Platynereis dumerilii (Lophotrochozoa). Conserved components of the pathway are identified and a scenario for their evolution in metazoans is proposed. Unexpectedly, neither Notch nor its ligands are expressed in the neurogenic epithelia of the larva at the time when massive neurogenesis begins. Using chemical inhibitors and neural markers, we demonstrate that Notch plays no major role in the general neurogenesis of larvae. Instead, we find Notch components expressed in nascent chaetal sacs, the organs that produce the annelid bristles. Impairing Notch signalling induces defects in chaetal sac formation, abnormalities in chaetae producing cells and a change of identity of chaeta growth accessory cells. This is the first bilaterian species in which the early neurogenesis processes appear to occur without a major involvement of the Notch pathway. Instead, Notch is co-opted to pattern annelid-specific organs, likely through a lateral inhibition process. These features reinforce the view that Notch signalling has been recruited multiple times in evolution due to its remarkable ‘toolkit’ nature.
Collapse
Affiliation(s)
- Eve Gazave
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| | - Quentin I B Lemaître
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| | - Guillaume Balavoine
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| |
Collapse
|
91
|
Fukusumi T, Guo TW, Sakai A, Ando M, Ren S, Haft S, Liu C, Amornphimoltham P, Gutkind JS, Califano JA. The NOTCH4- HEY1 Pathway Induces Epithelial-Mesenchymal Transition in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2017; 24:619-633. [PMID: 29146722 DOI: 10.1158/1078-0432.ccr-17-1366] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/16/2017] [Accepted: 11/09/2017] [Indexed: 12/26/2022]
Abstract
Purpose: Recently, several comprehensive genomic analyses demonstrated NOTCH1 and NOTCH3 mutations in head and neck squamous cell carcinoma (HNSCC) in approximately 20% of cases. Similar to other types of cancers, these studies also indicate that the NOTCH pathway is closely related to HNSCC progression. However, the role of NOTCH4 in HNSCC is less well understood.Experimental Design: We analyzed NOTCH4 pathway and downstream gene expression in the TCGA data set. To explore the functional role of NOTCH4, we performed in vitro proliferation, cisplatin viability, apoptosis, and cell-cycle assays. We also compared the relationships among NOTCH4, HEY1, and epithelial-mesenchymal transition (EMT)-related genes using the TCGA data set and in vitro assays.Results:HEY1 is specifically upregulated in HNSCC compared with normal tissues in the TCGA data set. NOTCH4 is more significantly related to HEY1 activation in HNSCC in comparison with other NOTCH receptors. NOTCH4 promotes cell proliferation, cisplatin resistance, inhibition of apoptosis, and cell-cycle dysregulation. Furthermore, NOTCH4 and HEY1 upregulation resulted in decreased E-cadherin expression and increased Vimentin, Fibronectin, TWIST1, and SOX2 expression. NOTCH4 and HEY1 expression was associated with an EMT phenotype as well as increased invasion and cell migration.Conclusions: In HNSCC, the NOTCH4-HEY1 pathway is specifically upregulated, induces proliferation and cisplatin resistance, and promotes EMT. Clin Cancer Res; 24(3); 619-33. ©2017 AACR.
Collapse
Affiliation(s)
- Takahito Fukusumi
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Theresa W Guo
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Akihiro Sakai
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Mizuo Ando
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Shuling Ren
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Sunny Haft
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Chao Liu
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Joseph A Califano
- Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
92
|
Pajcini KV, Xu L, Shao L, Petrovic J, Palasiewicz K, Ohtani Y, Bailis W, Lee C, Wertheim GB, Mani R, Muthusamy N, Li Y, Meijerink JPP, Blacklow SC, Faryabi RB, Cherry S, Pear WS. MAFB enhances oncogenic Notch signaling in T cell acute lymphoblastic leukemia. Sci Signal 2017; 10:10/505/eaam6846. [PMID: 29138297 DOI: 10.1126/scisignal.aam6846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Activating mutations in the gene encoding the cell-cell contact signaling protein Notch1 are common in human T cell acute lymphoblastic leukemias (T-ALLs). However, expressing Notch1 mutant alleles in mice fails to efficiently induce the development of leukemia. We performed a gain-of-function screen to identify proteins that enhanced signaling by leukemia-associated Notch1 mutants. The transcription factors MAFB and ETS2 emerged as candidates that individually enhanced Notch1 signaling, and when coexpressed, they synergistically increased signaling to an extent similar to that induced by core components of the Notch transcriptional complex. In mouse models of T-ALL, MAFB enhanced leukemogenesis by the naturally occurring Notch1 mutants, decreased disease latency, and increased disease penetrance. Decreasing MAFB abundance in mouse and human T-ALL cells reduced the expression of Notch1 target genes, including MYC and HES1, and sustained MAFB knockdown impaired T-ALL growth in a competitive setting. MAFB bound to ETS2 and interacted with the acetyltransferases PCAF and P300, highlighting its importance in recruiting coactivators that enhance Notch1 signaling. Together, these data identify a mechanism for enhancing the oncogenic potential of weak Notch1 mutants in leukemia models, and they reveal the MAFB-ETS2 transcriptional axis as a potential therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Kostandin V Pajcini
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA. .,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lanwei Xu
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lijian Shao
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jelena Petrovic
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karol Palasiewicz
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yumi Ohtani
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Will Bailis
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Curtis Lee
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajeswaran Mani
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Natarajan Muthusamy
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yunlei Li
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert B Faryabi
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
93
|
Choi SH, Severson E, Pear WS, Liu XS, Aster JC, Blacklow SC. The common oncogenomic program of NOTCH1 and NOTCH3 signaling in T-cell acute lymphoblastic leukemia. PLoS One 2017; 12:e0185762. [PMID: 29023469 PMCID: PMC5638296 DOI: 10.1371/journal.pone.0185762] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 09/19/2017] [Indexed: 11/24/2022] Open
Abstract
Notch is a major oncogenic driver in T cell acute lymphoblastic leukemia (T-ALL), in part because it binds to an enhancer that increases expression of MYC. Here, we exploit the capacity of activated NOTCH1 and NOTCH3 to induce T-ALL, despite substantial divergence in their intracellular regions, as a means to elucidate a broad, common Notch-dependent oncogenomic program through systematic comparison of the transcriptomes and Notch-bound genomic regulatory elements of NOTCH1- and NOTCH3-dependent T-ALL cells. ChIP-seq studies show a high concordance of functional NOTCH1 and NOTCH3 genomic binding sites that are enriched in binding motifs for RBPJ, the transcription factor that recruits activated Notch to DNA. The interchangeability of NOTCH1 and NOTCH3 was confirmed by rescue of NOTCH1-dependent T-ALL cells with activated NOTCH3 and vice versa. Despite remarkable overall similarity, there are nuanced differences in chromatin landscapes near critical common Notch target genes, most notably at a Notch-dependent enhancer that regulates MYC, which correlates with responsiveness to Notch pathway inhibitors. Overall, a common oncogenomic program driven by binding of either Notch is sufficient to maintain T-ALL cell growth, whereas cell-context specific differences appear to influence the response of T-ALL cells to Notch inhibition.
Collapse
Affiliation(s)
- Sung Hee Choi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, United States of America
| | - Eric Severson
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- Departments of Biostatistics and Computational Biology, Dana Farber Cancer Institute, and Harvard School of Public Health, Boston, MA, United States of America
| | - Warren S. Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Xiaole S. Liu
- Departments of Biostatistics and Computational Biology, Dana Farber Cancer Institute, and Harvard School of Public Health, Boston, MA, United States of America
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (SCB); (JCA)
| | - Stephen C. Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, United States of America
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (SCB); (JCA)
| |
Collapse
|
94
|
Notch-out for breast cancer therapies. N Biotechnol 2017; 39:215-221. [DOI: 10.1016/j.nbt.2017.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 07/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
|
95
|
Motooka Y, Fujino K, Sato Y, Kudoh S, Suzuki M, Ito T. Pathobiology of Notch2 in lung cancer. Pathology 2017; 49:486-493. [DOI: 10.1016/j.pathol.2017.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/03/2017] [Accepted: 05/08/2017] [Indexed: 12/29/2022]
|
96
|
Lachej N, Dabkevičienė D, Sasnauskienė A, Trimonytė RM, Kanopienė D, Kazbarienė B, Didžiapetrienė J. NOTCH signalinio kelio ir ginekologinių piktybinių navikų sąsaja. Acta Med Litu 2017. [PMID: 28630591 PMCID: PMC5467961 DOI: 10.6001/actamedica.v24i1.3461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Įvadas. Organizmo ląstelėse vykstančius procesus kontroliuoja įvairūs signaliniai keliai. Vienas iš jų yra NOTCH signalinis kelias. Nustatyta, kad dalinis NOTCH funkcijos praradimas arba nenormalus NOTCH signalo aktyvinimas susijęs su įvairiais žmogaus vystymosi sutrikimais ir ligomis. Medžiaga ir metodika. Pagrindinis informacijos šaltinis ieškant duomenų – PubMed duomenų bazė. Rezultatai. Straipsnyje nagrinėjama onkologinių ligų bei
NOTCH signalinio kelio dalyvių sąsaja. NOTCH signalas, vystantis vėžiui, gali veikti dvejopai: kaip onkogenas ir kaip naviko augimo slopiklis. Tikslus tokio poveikio mechanizmas dar nėra žinomas. NOTCH signalinio kelio tyrimai svarbūs siekiant atrasti naujus vėžio gydymo būdus, farmakologiniais ir genetiniais metodais valdant NOTCH signalinį kelią. Šioje apžvalgoje daugiausia dėmesio skiriama ginekologiniams piktybiniams navikams, ypač gimdos kūno vėžiui. Išvados. Pastarųjų metų mokslinių tyrimų duomenys rodo, kad NOTCH signalinis kelias yra neabejotinai svarbus formuojantis gimdos kūno vėžiui, todėl jo komponentai gali būti potencialūs prognoziniai biožymenys ir molekuliniai terapiniai taikiniai. Siekiant patikslinti NOTCH signalinio kelio dalyvių reikšmę bei jų sąveiką su kitų signalinių kelių dalyviais, kurie taip pat gali būti svarbūs formuojantis ir progresuojant gimdos kūno vėžiui, reikalingi tolesni šios srities moksliniai tyrimai.
Collapse
Affiliation(s)
| | - Daiva Dabkevičienė
- Vilniaus universitetas, Gamtos mokslų fakultetas, Biochemijos ir molekulinės biologijos katedra
| | - Aušra Sasnauskienė
- Vilniaus universitetas, Gamtos mokslų fakultetas, Biochemijos ir molekulinės biologijos katedra
| | - Rūta Marija Trimonytė
- Vilniaus universitetas, Gamtos mokslų fakultetas, Biochemijos ir molekulinės biologijos katedra
| | | | | | | |
Collapse
|
97
|
Han X, Ranganathan P, Tzimas C, Weaver KL, Jin K, Astudillo L, Zhou W, Zhu X, Li B, Robbins DJ, Capobianco AJ. Notch Represses Transcription by PRC2 Recruitment to the Ternary Complex. Mol Cancer Res 2017; 15:1173-1183. [PMID: 28584023 DOI: 10.1158/1541-7786.mcr-17-0241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 11/16/2022]
Abstract
It is well established that Notch functions as a transcriptional activator through the formation of a ternary complex that comprises Notch, Maml, and CSL. This ternary complex then serves to recruit additional transcriptional cofactors that link to higher order transcriptional complexes. The mechanistic details of these events remain unclear. This report reveals that the Notch ternary complex can direct the formation of a repressor complex to terminate gene expression of select target genes. Herein, it is demonstrated that p19Arf and Klf4 are transcriptionally repressed in a Notch-dependent manner. Furthermore, results indicate that Notch recruits Polycomb Repressor Complex 2 (PRC2) and Lysine Demethylase 1 (KDM1A/LSD1) to these promoters, which leads to changes in the epigenetic landscape and repression of transcription. The demethylase activity of LSD1 is a prerequisite for Notch-mediated transcriptional repression. In addition, a stable Notch transcriptional repressor complex was identified containing LSD1, PRC2, and the Notch ternary complex. These findings demonstrate a novel function of Notch and provide further insight into the mechanisms of Notch-mediated tumorigenesis.Implications: This study provides rationale for the targeting of epigenetic enzymes to inhibit Notch activity or use in combinatorial therapy to provide a more profound therapeutic response. Mol Cancer Res; 15(9); 1173-83. ©2017 AACR.
Collapse
Affiliation(s)
- Xiaoqing Han
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.,The Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Prathibha Ranganathan
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.,Centre for Human Genetics, Electronic City, Bengaluru, Karnataka, India
| | - Christos Tzimas
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Kelly L Weaver
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Ke Jin
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.,The Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Luisana Astudillo
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Wen Zhou
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xiaoxia Zhu
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Bin Li
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - David J Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
98
|
Kimball AS, Joshi AD, Boniakowski AE, Schaller M, Chung J, Allen R, Bermick J, Carson WF, Henke PK, Maillard I, Kunkel SL, Gallagher KA. Notch Regulates Macrophage-Mediated Inflammation in Diabetic Wound Healing. Front Immunol 2017; 8:635. [PMID: 28620387 PMCID: PMC5451506 DOI: 10.3389/fimmu.2017.00635] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022] Open
Abstract
Macrophages are essential immune cells necessary for regulated inflammation during wound healing. Recent studies have identified that Notch plays a role in macrophage-mediated inflammation. Thus, we investigated the role of Notch signaling on wound macrophage phenotype and function during normal and diabetic wound healing. We found that Notch receptor and ligand expression are dynamic in wound macrophages during normal healing. Mice with a myeloid-specific Notch signaling defect (DNMAMLfloxedLyz2Cre+) demonstrated delayed early healing (days 1–3) and wound macrophages had decreased inflammatory gene expression. In our physiologic murine model of type 2 diabetes (T2D), Notch receptor expression was significantly increased in wound macrophages on day 6, following the initial inflammatory phase of wound healing, corresponding to increased inflammatory cytokine expression. This increase in Notch1 and Notch2 was also observed in human monocytes from patients with T2D. Further, in prediabetic mice with a genetic Notch signaling defect (DNMAMLfloxedLyz2Cre+ on a high-fat diet), improved wound healing was seen at late time points (days 6–7). These findings suggest that Notch is critical for the early inflammatory phase of wound healing and directs production of macrophage-dependent inflammatory mediators. These results identify that canonical Notch signaling is important in directing macrophage function in wound repair and define a translational target for the treatment of non-healing diabetic wounds.
Collapse
Affiliation(s)
- Andrew S Kimball
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Amrita D Joshi
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Anna E Boniakowski
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Matthew Schaller
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Jooho Chung
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Ronald Allen
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer Bermick
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - William F Carson
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Peter K Henke
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Ivan Maillard
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Steve L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
99
|
Guest RV, Boulter L, Dwyer BJ, Forbes SJ. Understanding liver regeneration to bring new insights to the mechanisms driving cholangiocarcinoma. NPJ Regen Med 2017; 2:13. [PMID: 29302349 PMCID: PMC5677951 DOI: 10.1038/s41536-017-0018-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/22/2017] [Accepted: 03/06/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer frequently arises in epithelial tissues subjected to repeated cycles of injury and repair. Improving our understanding of tissue regeneration is, therefore, likely to reveal novel processes with inherent potential for aberration that can lead to carcinoma. These highly conserved regenerative mechanisms are increasingly understood and in the liver are associated with special characteristics that underlie the organ's legendary capacity for restoration of size and function following even severe or chronic injury. The nature of the injury can determine the cellular source of epithelial regeneration and the signalling mechanisms brought to play. These observations are shaping how we understand and experimentally investigate primary liver cancer, in particular cholangiocarcinoma; a highly invasive malignancy of the bile ducts, resistant to chemotherapy and whose pathogenesis has hitherto been poorly understood. Interestingly, signals that drive liver development become activated in the formation of cholangiocarcinoma, such as Notch and Wnt and may be potential future therapeutic targets. In this review, we summarise the work which has led to the current understanding of the cellular source of cholangiocarcinoma, how the tumour recruits, sustains and is educated by its supporting stromal environment, and the tumour-derived signals that drive the progression and invasion of the cancer. With few current treatments of any true efficacy, advances that will improve our understanding of the mechanisms driving this aggressive malignancy are welcome and may help drive therapeutic developments.
Collapse
Affiliation(s)
- R. V. Guest
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, , Edinburgh, EH16 4UU UK
| | - L. Boulter
- Institute for Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, , Edinburgh, EH4 2XU UK
| | - B. J. Dwyer
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, , Edinburgh, EH16 4UU UK
| | - S. J. Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, , Edinburgh, EH16 4UU UK
| |
Collapse
|
100
|
The role of Notch signaling in gastric carcinoma: molecular pathogenesis and novel therapeutic targets. Oncotarget 2017; 8:53839-53853. [PMID: 28881855 PMCID: PMC5581154 DOI: 10.18632/oncotarget.17809] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/17/2017] [Indexed: 12/14/2022] Open
Abstract
Notch signaling, an evolutionarily conserved signaling cascade system, is involved in promoting the progression of different types of cancers. Within the past decades, the Notch signaling pathway has increasingly been shown to have a primary role in deciding the fate of cancer cells and cancer stem cells in the stomach. Most components of Notch signaling are strongly expressed at different levels in gastric carcinoma tissue samples and are associated with a considerable number of clinical parameters. Moreover, crosstalk signaling between the Notch pathway and the Wnt, Ras, and NF-κB pathways promotes the process of gastric carcinogenesis. Consequently, this increases proliferation and prevents apoptosis in gastric cancer cells, and it contributes to the induction of angiogenesis and accelerates the progression of the epithelial-to-mesenchymal transition. Although the Notch signaling pathway presents novel therapeutic targets for cancer therapeutic intervention, there is still a dearth of in-depth understanding of the molecular mechanisms of Notch signaling in gastric carcinoma. In this review, we summarize the landscape of the Notch signaling pathway and recent findings on Notch signaling in gastric cancer. Furthermore, advanced studies and clinical treatments targeting the Notch signaling pathway arediscussed.
Collapse
|