201
|
Abstract
Notch signaling is an evolutionarily conserved pathway that is essential for development, where it controls processes ranging from cell differentiation to survival. Transport through endosomes is a critical step in regulating Notch signaling capacity, where the E3 ubiquitin ligase DTX1 is thought to control Notch1 intracellular transport decisions by direct receptor ubiquitination. However, how DTX1 regulates Notch1 transport within endosomes and the consequence of Notch1 ubiquitination by DTX1 remain unresolved. Here we demonstrate that DTX1 colocalizes with Notch1 on tubulovesicular recycling endosomes. We find that DTX1 silencing leads to enhanced Notch1 recycling from this compartment to the cell surface via a rab4a-mediated transport route. This, in turn, increases Notch1 cell-surface levels and enhances signaling. Surprisingly, we discovered that DTX1 depletion also elevates Notch1 activity mediated by a mutant form of the receptor that lacks lysine residues for ubiquitination, suggesting that DTX1 targets additional factors. Using an activity-based screen for ubiquitination targets, we identified multiple DTX1 substrates including PI5P4Kγ, a lipid kinase involved in PI(4,5)P2 production. Immunolocalization analysis reveals that PI5P4Kγ, like DTX1 and Notch1, is present on tubulovesicular recycling endosomes. However, in contrast to DTX1, Notch1 signaling is inhibited by pharmacological inactivation or siRNA depletion of PI5P4Kγ. Moreover, loss of PI5P4Kγ activity decreases Notch1 recycling rates and reduces receptor cell-surface levels. Collectively, these findings argue that PI5P4Kγ positively regulates the Notch pathway by promoting receptor recycling. Additionally, they support a model where DTX1 controls Notch1 endosomal sorting decisions by controlling PI5P4Kγ-mediated production of PI(4,5)P2.
Collapse
Affiliation(s)
- Li Zheng
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| | - Sean D Conner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
202
|
Kim J, Bilder D, Neufeld TP. Mechanical stress regulates insulin sensitivity through integrin-dependent control of insulin receptor localization. Genes Dev 2018; 32:156-164. [PMID: 29440263 PMCID: PMC5830928 DOI: 10.1101/gad.305870.117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/02/2018] [Indexed: 12/22/2022]
Abstract
Kim et al. show that insulin signaling in Drosophila adipocytes is abolished in the absence of physical activity and mechanical stress. The insulin receptor and downstream components are recruited to the plasma membrane upon stress sensing mediated by integrins. Insulin resistance, the failure to activate insulin signaling in the presence of ligand, leads to metabolic diseases, including type 2 diabetes. Physical activity and mechanical stress have been shown to protect against insulin resistance, but the molecular mechanisms remain unclear. Here, we address this relationship in the Drosophila larval fat body, an insulin-sensitive organ analogous to vertebrate adipose tissue and livers. We found that insulin signaling in Drosophila fat body cells is abolished in the absence of physical activity and mechanical stress even when excess insulin is present. Physical movement is required for insulin sensitivity in both intact larvae and fat bodies cultured ex vivo. Interestingly, the insulin receptor and other downstream components are recruited to the plasma membrane in response to mechanical stress, and this membrane localization is rapidly lost upon disruption of larval or tissue movement. Sensing of mechanical stimuli is mediated in part by integrins, whose activation is necessary and sufficient for mechanical stress-dependent insulin signaling. Insulin resistance develops naturally during the transition from the active larval stage to the immotile pupal stage, suggesting that regulation of insulin sensitivity by mechanical stress may help coordinate developmental programming with metabolism.
Collapse
Affiliation(s)
- Jung Kim
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
203
|
Rewiring T-cell responses to soluble factors with chimeric antigen receptors. Nat Chem Biol 2018; 14:317-324. [PMID: 29377003 PMCID: PMC6035732 DOI: 10.1038/nchembio.2565] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/12/2017] [Indexed: 12/22/2022]
Abstract
Chimeric antigen receptor (CAR)-expressing T cells targeting surface-bound tumor antigens have yielded promising clinical outcomes, with two CD19 CAR-T cell therapies recently receiving FDA approval for the treatment of B-cell malignancies. The adoption of CARs for the recognition of soluble ligands, a distinct class of biomarkers in physiology and disease, could considerably broaden the utility of CARs in disease treatment. In this study, we demonstrate that CAR-T cells can be engineered to respond robustly to diverse soluble ligands, including the CD19 ectodomain, GFP variants, and transforming growth factor beta (TGF-β). We additionally show that CAR signaling in response to soluble ligands relies on ligand-mediated CAR dimerization and that CAR responsiveness to soluble ligands can be fine-tuned by adjusting the mechanical coupling between the CAR's ligand-binding and signaling domains. Our results support a role for mechanotransduction in CAR signaling and demonstrate an approach for systematically engineering immune-cell responses to soluble, extracellular ligands.
Collapse
|
204
|
Roybal KT, Lim WA. Synthetic Immunology: Hacking Immune Cells to Expand Their Therapeutic Capabilities. Annu Rev Immunol 2018; 35:229-253. [PMID: 28446063 DOI: 10.1146/annurev-immunol-051116-052302] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The ability of immune cells to survey tissues and sense pathologic insults and deviations makes them a unique platform for interfacing with the body and disease. With the rapid advancement of synthetic biology, we can now engineer and equip immune cells with new sensors and controllable therapeutic response programs to sense and treat diseases that our natural immune system cannot normally handle. Here we review the current state of engineered immune cell therapeutics and their unique capabilities compared to small molecules and biologics. We then discuss how engineered immune cells are being designed to combat cancer, focusing on how new synthetic biology tools are providing potential ways to overcome the major roadblocks for treatment. Finally, we give a long-term vision for the use of synthetic biology to engineer immune cells as a general sensor-response platform to precisely detect disease, to remodel disease microenvironments, and to treat a potentially wide range of challenging diseases.
Collapse
Affiliation(s)
- Kole T Roybal
- Parker Institute for Cancer Immunotherapy, Department of Microbiology and Immunology, University of California, San Francisco, California 94143;
| | - Wendell A Lim
- Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158;
| |
Collapse
|
205
|
Tschumperlin DJ, Ligresti G, Hilscher MB, Shah VH. Mechanosensing and fibrosis. J Clin Invest 2018; 128:74-84. [PMID: 29293092 DOI: 10.1172/jci93561] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue injury disrupts the mechanical homeostasis that underlies normal tissue architecture and function. The failure to resolve injury and restore homeostasis gives rise to progressive fibrosis that is accompanied by persistent alterations in the mechanical environment as a consequence of pathological matrix deposition and stiffening. This Review focuses on our rapidly growing understanding of the molecular mechanisms linking the altered mechanical environment in injury, repair, and fibrosis to cellular activation. In particular, our focus is on the mechanisms by which cells transduce mechanical signals, leading to transcriptional and epigenetic responses that underlie both transient and persistent alterations in cell state that contribute to fibrosis. Translation of these mechanobiological insights may enable new approaches to promote tissue repair and arrest or reverse fibrotic tissue remodeling.
Collapse
Affiliation(s)
| | | | - Moira B Hilscher
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
206
|
Control of Blood Vessel Formation by Notch Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:319-338. [PMID: 30030834 DOI: 10.1007/978-3-319-89512-3_16] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Blood vessels span throughout the body to nourish tissue cells and to provide gateways for immune surveillance. Endothelial cells that line capillaries have the remarkable capacity to be quiescent for years but to switch rapidly into the activated state once new blood vessels need to be formed. In addition, endothelial cells generate niches for progenitor and tumor cells and provide organ-specific paracrine (angiocrine) factors that control organ development and regeneration, maintenance of homeostasis and tumor progression. Recent data indicate a pivotal role for blood vessels in responding to metabolic changes and that endothelial cell metabolism is a novel regulator of angiogenesis. The Notch pathway is the central signaling mode that cooperates with VEGF, WNT, BMP, TGF-β, angiopoietin signaling and cell metabolism to orchestrate angiogenesis, tip/stalk cell selection and arteriovenous specification. Here, we summarize the current knowledge and implications regarding the complex roles of Notch signaling during physiological and tumor angiogenesis, the dynamic nature of tip/stalk cell selection in the nascent vessel sprout and arteriovenous differentiation. Furthermore, we shed light on recent work on endothelial cell metabolism, perfusion-independent angiocrine functions of endothelial cells in organ-specific vascular beds and how manipulation of Notch signaling may be used to target the tumor vasculature.
Collapse
|
207
|
Lovendahl KN, Blacklow SC, Gordon WR. The Molecular Mechanism of Notch Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:47-58. [PMID: 30030821 DOI: 10.1007/978-3-319-89512-3_3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Research in the last several years has shown that Notch proteolysis, and thus Notch activation, is conformationally controlled by the extracellular juxtamembrane NRR of Notch, which sterically occludes the S2 protease site until ligand binds. The question of how conformational exposure of the protease site is achieved during physiologic activation, and thus how normal activation is bypassed in disease pathogenesis, has been the subject of intense study in the last several years, and is the subject of this chapter. Here, we summarize the structural features of the NRR domains of Notch receptors that establish the autoinhibited state and then review a number of recent studies aimed at testing the mechanotransduction model for Notch signaling using force spectroscopy and molecular tension sensors.
Collapse
Affiliation(s)
- Klaus N Lovendahl
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
208
|
CSL-Associated Corepressor and Coactivator Complexes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:279-295. [PMID: 30030832 DOI: 10.1007/978-3-319-89512-3_14] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The highly conserved Notch signal transduction pathway orchestrates fundamental cellular processes including, differentiation, proliferation, and apoptosis during embryonic development and in the adult organism. Dysregulated Notch signaling underlies the etiology of a variety of human diseases, such as certain types of cancers, developmental disorders and cardiovascular disease. Ligand binding induces proteolytic cleavage of the Notch receptor and nuclear translocation of the Notch intracellular domain (NICD), which forms a ternary complex with the transcription factor CSL and the coactivator MAML to upregulate transcription of Notch target genes. The DNA-binding protein CSL is the centrepiece of transcriptional regulation in the Notch pathway, acting as a molecular hub for interactions with either corepressors or coactivators to repress or activate, respectively, transcription. Here we review previous structure-function studies of CSL-associated coregulator complexes and discuss the molecular insights gleaned from this research. We discuss the functional consequences of both activating and repressing binding partners using the same interaction platforms on CSL. We also emphasize that although there has been a significant uptick in structural information over the past decade, it is still under debate how the molecular switch from repression to activation mediated by CSL occurs at Notch target genes and whether it will be possible to manipulate these transcription complexes therapeutically in the future.
Collapse
|
209
|
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
|
210
|
Fang JS, Coon BG, Gillis N, Chen Z, Qiu J, Chittenden TW, Burt JM, Schwartz MA, Hirschi KK. Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat Commun 2017; 8:2149. [PMID: 29247167 PMCID: PMC5732288 DOI: 10.1038/s41467-017-01742-7] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 10/13/2017] [Indexed: 01/26/2023] Open
Abstract
Establishment of a functional vascular network is rate-limiting in embryonic development, tissue repair and engineering. During blood vessel formation, newly generated endothelial cells rapidly expand into primitive plexi that undergo vascular remodeling into circulatory networks, requiring coordinated growth inhibition and arterial-venous specification. Whether the mechanisms controlling endothelial cell cycle arrest and acquisition of specialized phenotypes are interdependent is unknown. Here we demonstrate that fluid shear stress, at arterial flow magnitudes, maximally activates NOTCH signaling, which upregulates GJA4 (commonly, Cx37) and downstream cell cycle inhibitor CDKN1B (p27). Blockade of any of these steps causes hyperproliferation and loss of arterial specification. Re-expression of GJA4 or CDKN1B, or chemical cell cycle inhibition, restores endothelial growth control and arterial gene expression. Thus, we elucidate a mechanochemical pathway in which arterial shear activates a NOTCH-GJA4-CDKN1B axis that promotes endothelial cell cycle arrest to enable arterial gene expression. These insights will guide vascular regeneration and engineering.
Collapse
Affiliation(s)
- Jennifer S Fang
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Brian G Coon
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Noelle Gillis
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Zehua Chen
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE 55 Cambridge Parkway, 8th Floor, Cambridge, MA, 02142, USA
| | - Jingyao Qiu
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Thomas W Chittenden
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE 55 Cambridge Parkway, 8th Floor, Cambridge, MA, 02142, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, A-111, 25 Shattuck Street, Boston, MA, 02115, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street #56-651, Cambridge, MA, 02142, USA
| | - Janis M Burt
- Department of Physiology, College of Medicine, The University of Arizona, 1501 N. Campbell Road, Tucson, AZ, 85724, USA
| | - Martin A Schwartz
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Karen K Hirschi
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
211
|
Huang TH, Niesman P, Arasu D, Lee D, De La Cruz AL, Callejas A, Hong EJ, Lois C. Tracing neuronal circuits in transgenic animals by transneuronal control of transcription ( TRACT). eLife 2017; 6:32027. [PMID: 29231171 PMCID: PMC5777821 DOI: 10.7554/elife.32027] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/02/2017] [Indexed: 12/20/2022] Open
Abstract
Understanding the computations that take place in brain circuits requires identifying how neurons in those circuits are connected to one another. We describe a technique called TRACT (TRAnsneuronal Control of Transcription) based on ligand-induced intramembrane proteolysis to reveal monosynaptic connections arising from genetically labeled neurons of interest. In this strategy, neurons expressing an artificial ligand (‘donor’ neurons) bind to and activate a genetically-engineered artificial receptor on their synaptic partners (‘receiver’ neurons). Upon ligand-receptor binding at synapses the receptor is cleaved in its transmembrane domain and releases a protein fragment that activates transcription in the synaptic partners. Using TRACT in Drosophila we have confirmed the connectivity between olfactory receptor neurons and their postsynaptic targets, and have discovered potential new connections between neurons in the circadian circuit. Our results demonstrate that the TRACT method can be used to investigate the connectivity of neuronal circuits in the brain. One of the main obstacles to understanding how the brain works is that we know relatively little about how its nerve cells or neurons are connected to one another. These connections make up the brain’s wiring diagram. Current methods for revealing this wiring all have limitations. The most popular method – serial electron microscopy – can reveal the connections in a small region of the brain in great detail, but it cannot show connections between neurons that are far apart. Huang et al. have now created a genetic system for visualizing these connections. For neurons to communicate, one neuron must produce a signal called a ligand. This ligand can then bind to and activate its partner neuron. Huang et al. modified the DNA of neurons so that every time those cells produced a specific ligand, they also produced a red fluorescent protein. Similar modifications ensured that every time the ligand activated a partner neuron, the activated neuron produced a green fluorescent protein. Viewing the red and green neurons under a microscope enabled Huang et al. to see which cells were communicating with which others. While these experiments took place in fruit flies, the same approach should also work in other laboratory animals, including fish, mice and rats. Once we know the wiring diagram of the brain, the next step is to investigate the role of the various connections. To understand how a computer works, for example, we might change the connections between its circuit components and look at how this affects the computer’s output. With this new method, we can change how neurons communicate with one another in the brain, and then look at the effects on behavior. This should provide insights into the workings of the human brain, and clues to what goes wrong in disorders like schizophrenia and autism.
Collapse
Affiliation(s)
- Ting-Hao Huang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Peter Niesman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Deepshika Arasu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Donghyung Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Aubrie L De La Cruz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Antuca Callejas
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States.,Department of Cell Biology, School of Science, University of Extremadura, Badajoz, Spain
| | - Elizabeth J Hong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
212
|
Xiao Z, Baudry J, Cao L, Huang J, Chen H, Yates CR, Li W, Dong B, Waters CM, Smith JC, Quarles LD. Polycystin-1 interacts with TAZ to stimulate osteoblastogenesis and inhibit adipogenesis. J Clin Invest 2017; 128:157-174. [PMID: 29202470 DOI: 10.1172/jci93725] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/17/2017] [Indexed: 01/15/2023] Open
Abstract
The molecular mechanisms that transduce the osteoblast response to physical forces in the bone microenvironment are poorly understood. Here, we used genetic and pharmacological experiments to determine whether the polycystins PC1 and PC2 (encoded by Pkd1 and Pkd2) and the transcriptional coactivator TAZ form a mechanosensing complex in osteoblasts. Compound-heterozygous mice lacking 1 copy of Pkd1 and Taz exhibited additive decrements in bone mass, impaired osteoblast-mediated bone formation, and enhanced bone marrow fat accumulation. Bone marrow stromal cells and osteoblasts derived from these mice showed impaired osteoblastogenesis and enhanced adipogenesis. Increased extracellular matrix stiffness and application of mechanical stretch to multipotent mesenchymal cells stimulated the nuclear translocation of the PC1 C-terminal tail/TAZ (PC1-CTT/TAZ) complex, leading to increased runt-related transcription factor 2-mediated (Runx2-mediated) osteogenic and decreased PPARγ-dependent adipogenic gene expression. Using structure-based virtual screening, we identified a compound predicted to bind to PC2 in the PC1:PC2 C-terminal tail region with helix:helix interaction. This molecule stimulated polycystin- and TAZ-dependent osteoblastogenesis and inhibited adipogenesis. Thus, we show that polycystins and TAZ integrate at the molecular level to reciprocally regulate osteoblast and adipocyte differentiation, indicating that the polycystins/TAZ complex may be a potential therapeutic target to increase bone mass.
Collapse
Affiliation(s)
- Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jerome Baudry
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Li Cao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jinsong Huang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Hao Chen
- Department of Pharmaceutical Sciences and
| | | | - Wei Li
- Department of Pharmaceutical Sciences and
| | - Brittany Dong
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christopher M Waters
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeremy C Smith
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
213
|
A non-canonical Notch complex regulates adherens junctions and vascular barrier function. Nature 2017; 552:258-262. [PMID: 29160307 PMCID: PMC5730479 DOI: 10.1038/nature24998] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/06/2017] [Indexed: 01/01/2023]
Abstract
The vascular barrier that separates blood from tissues is actively regulated by the endothelium and is essential for transport, inflammation, and haemostasis. Haemodynamic shear stress plays a critical role in maintaining endothelial barrier function, but how this occurs remains unknown. Here we use an engineered organotypic model of perfused microvessels to show that activation of the transmembrane receptor NOTCH1 directly regulates vascular barrier function through a non-canonical, transcription-independent signalling mechanism that drives assembly of adherens junctions, and confirm these findings in mouse models. Shear stress triggers DLL4-dependent proteolytic activation of NOTCH1 to expose the transmembrane domain of NOTCH1. This domain mediates establishment of the endothelial barrier; expression of the transmembrane domain of NOTCH1 is sufficient to rescue defects in barrier function induced by knockout of NOTCH1. The transmembrane domain restores barrier function by catalysing the formation of a receptor complex in the plasma membrane consisting of vascular endothelial cadherin, the transmembrane protein tyrosine phosphatase LAR, and the RAC1 guanidine-exchange factor TRIO. This complex activates RAC1 to drive assembly of adherens junctions and establish barrier function. Canonical transcriptional signalling via Notch is highly conserved in metazoans and is required for many processes in vascular development, including arterial-venous differentiation, angiogenesis and remodelling. We establish the existence of a non-canonical cortical NOTCH1 signalling pathway that regulates vascular barrier function, and thus provide a mechanism by which a single receptor might link transcriptional programs with adhesive and cytoskeletal remodelling.
Collapse
|
214
|
Martin JS, Kephart WC, Haun CT, McCloskey AE, Shake JJ, Mobley CB, Goodlett MD, Kavazis A, Pascoe DD, Zhang L, Roberts MD. Impact of external pneumatic compression target inflation pressure on transcriptome-wide RNA expression in skeletal muscle. Physiol Rep 2017; 4:4/22/e13029. [PMID: 27884954 PMCID: PMC5357997 DOI: 10.14814/phy2.13029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/03/2016] [Accepted: 10/12/2016] [Indexed: 11/24/2022] Open
Abstract
Next‐generation RNA sequencing was employed to determine the acute and subchronic impact of peristaltic pulse external pneumatic compression (PEPC) of different target inflation pressures on global gene expression in human vastus lateralis skeletal muscle biopsy samples. Eighteen (N = 18) male participants were randomly assigned to one of the three groups: (1) sham (n = 6), 2) EPC at 30–40 mmHg (LP‐EPC; n = 6), and 3) EPC at 70–80 mmHg (MP‐EPC; n = 6). One hour treatment with sham/EPC occurred for seven consecutive days. Vastus lateralis skeletal muscle biopsies were performed at baseline (before first treatment; PRE), 1 h following the first treatment (POST1), and 24 h following the last (7th) treatment (POST2). Changes from PRE in gene expression were analyzed via paired comparisons within each group. Genes were filtered to include only those that had an RPKM ≥ 1.0, a fold‐change of ≥1.5 and a paired t‐test value of <0.01. For the sham condition, two genes at POST1 and one gene at POST2 were significantly altered. For the LP‐EPC condition, nine genes were up‐regulated and 0 genes were down‐regulated at POST1 while 39 genes were up‐regulated and one gene down‐regulated at POST2. For the MP‐EPC condition, two genes were significantly up‐regulated and 21 genes were down‐regulated at POST1 and 0 genes were altered at POST2. Both LP‐EPC and MP‐EPC acutely alter skeletal muscle gene expression, though only LP‐EPC appeared to affect gene expression with subchronic application. Moreover, the transcriptome response to EPC demonstrated marked heterogeneity (i.e., genes and directionality) with different target inflation pressures.
Collapse
Affiliation(s)
- Jeffrey S Martin
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama .,School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Cody T Haun
- School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Joshua J Shake
- School of Kinesiology, Auburn University, Auburn, Alabama
| | | | - Michael D Goodlett
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,Athletics Department, Auburn University, Auburn, Alabama
| | - Andreas Kavazis
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,School of Kinesiology, Auburn University, Auburn, Alabama
| | - David D Pascoe
- School of Kinesiology, Auburn University, Auburn, Alabama
| | - Lee Zhang
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama
| | - Michael D Roberts
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine - Auburn Campus, Auburn, Alabama.,School of Kinesiology, Auburn University, Auburn, Alabama
| |
Collapse
|
215
|
Chromosome Intermingling: Mechanical Hotspots for Genome Regulation. Trends Cell Biol 2017; 27:810-819. [DOI: 10.1016/j.tcb.2017.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/20/2017] [Accepted: 06/20/2017] [Indexed: 11/20/2022]
|
216
|
Sjöqvist M, Andersson ER. Do as I say, Not(ch) as I do: Lateral control of cell fate. Dev Biol 2017; 447:58-70. [PMID: 28969930 DOI: 10.1016/j.ydbio.2017.09.032] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 01/19/2023]
Abstract
Breaking symmetry in populations of uniform cells, to induce adoption of an alternative cell fate, is an essential developmental mechanism. Similarly, domain and boundary establishment are crucial steps to forming organs during development. Notch signaling is a pathway ideally suited to mediating precise patterning cues, as both receptors and ligands are membrane-bound and can thus act as a precise switch to toggle cell fates on or off. Fine-tuning of signaling by positive or negative feedback mechanisms dictate whether signaling results in lateral induction or lateral inhibition, respectively, allowing Notch to either induce entire regions of cell specification, or dictate binary fate choices. Furthermore, pathway activity is modulated by Fringe modification of receptors or ligands, co-expression of receptors with ligands, mode of ligand presentation, and cell surface area in contact. In this review, we describe how Notch signaling is fine-tuned to mediate lateral induction or lateral inhibition cues, and discuss examples from C.elegans, D. melanogaster and M. musculus. Identifying the cellular machinery dictating the choice between lateral induction and lateral inhibition highlights the versatility of the Notch signaling pathway in development.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Emma R Andersson
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden.
| |
Collapse
|
217
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 686] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
218
|
Perez-Mockus G, Schweisguth F. Cell Polarity and Notch Signaling: Linked by the E3 Ubiquitin Ligase Neuralized? Bioessays 2017; 39. [DOI: 10.1002/bies.201700128] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/17/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Gantas Perez-Mockus
- Institut Pasteur,; Dept of Developmental and Stem Cell Biology; F-75015 Paris France
- CNRS; UMR3738; F-75015 Paris France
- Univ. Pierre et Marie Curie; Cellule Pasteur UPMC; rue du Dr Roux 75015 Paris France
| | - Francois Schweisguth
- Institut Pasteur,; Dept of Developmental and Stem Cell Biology; F-75015 Paris France
- CNRS; UMR3738; F-75015 Paris France
| |
Collapse
|
219
|
The Canonical Notch Signaling Pathway: Structural and Biochemical Insights into Shape, Sugar, and Force. Dev Cell 2017; 41:228-241. [PMID: 28486129 DOI: 10.1016/j.devcel.2017.04.001] [Citation(s) in RCA: 290] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/04/2017] [Accepted: 04/03/2017] [Indexed: 02/07/2023]
Abstract
The Notch signaling pathway relies on a proteolytic cascade to release its transcriptionally active intracellular domain, on force to unfold a protective domain and permit proteolysis, on extracellular domain glycosylation to tune the forces exerted by endocytosed ligands, and on a motley crew of nuclear proteins, chromatin modifiers, ubiquitin ligases, and a few kinases to regulate activity and half-life. Herein we provide a review of recent molecular insights into how Notch signals are triggered and how cell shape affects these events, and we use the new insights to illuminate a few perplexing observations.
Collapse
|
220
|
Abstract
Human development requires intricate cell specification and communication pathways that allow an embryo to generate and appropriately connect more than 200 different cell types. Key to the successful completion of this differentiation programme is the quantitative and reversible regulation of core signalling networks, and post-translational modification with ubiquitin provides embryos with an essential tool to accomplish this task. Instigated by E3 ligases and reversed by deubiquitylases, ubiquitylation controls many processes that are fundamental for development, such as cell division, fate specification and migration. As aberrant function or regulation of ubiquitylation enzymes is at the roots of developmental disorders, cancer, and neurodegeneration, modulating the activity of ubiquitylation enzymes is likely to provide strategies for therapeutic intervention.
Collapse
|
221
|
Multiplexing molecular tension sensors reveals piconewton force gradient across talin-1. Nat Methods 2017; 14:1090-1096. [DOI: 10.1038/nmeth.4431] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/18/2017] [Indexed: 01/09/2023]
|
222
|
Safaee H, Bakooshli MA, Davoudi S, Cheng RY, Martowirogo AJ, Li EW, Simmons CA, Gilbert PM. Tethered Jagged-1 Synergizes with Culture Substrate Stiffness to Modulate Notch-Induced Myogenic Progenitor Differentiation. Cell Mol Bioeng 2017; 10:501-513. [PMID: 31719873 DOI: 10.1007/s12195-017-0506-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/17/2017] [Indexed: 12/19/2022] Open
Abstract
Introduction Notch signaling is amongst the key intrinsic mechanisms regulating satellite cell fate, promoting the transition of activated satellite cells to highly proliferative myogenic progenitor cells and preventing their premature differentiation. Although much is known about the biochemical milieu that drives myogenic progression, less is known about the spatial cues providing spatiotemporal control of skeletal muscle repair in the context of Notch signaling. Methods Using a murine injury model, we quantified in vivo biophysical changes that occur within the skeletal muscle during regeneration. Employing tunable poly(ethylene glycol)-based hydrogel substrates, we modeled the measured changes in bulk stiffness in the context of Notch ligand signaling, which are present in the regenerative milieu at the time of injury. Results Following injury, there is a transient increase in the bulk stiffness of the tibialis anterior muscle that may be explained in part by changes in extracellular matrix deposition. When presented to primary myoblasts, Jagged-1, Jagged-2, and Dll1 in a tethered format elicited greater degrees of Notch activity compared to their soluble form. Only tethered Jagged-1 effects were tuned by substrate stiffness, with the greatest Notch activation observed on stiff hydrogels matching the stiffness of regenerating muscle. When exposed to tethered Jagged-1 on stiff hydrogels, fewer primary myoblasts expressed myogenin, and pharmacological inhibitor studies suggest this effect is Notch and RhoA dependent. Conclusion Our study proposes that tethered Jagged-1 presented in the context of transient tissue stiffening serves to tune Notch activity in myogenic progenitors during skeletal muscle repair and delay differentiation.
Collapse
Affiliation(s)
- Helia Safaee
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Mohsen A Bakooshli
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Sadegh Davoudi
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Richard Y Cheng
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Aditya J Martowirogo
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Edward W Li
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Craig A Simmons
- 1Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1 Canada.,Translational Biology and Engineering Program, The Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1 Canada.,3Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Penney M Gilbert
- 4Department of Biochemistry, University of Toronto, Toronto, ON Canada.,5Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON Canada.,6Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Rm. 407, Toronto, ON M5S 3E1 Canada
| |
Collapse
|
223
|
Baratchi S, Khoshmanesh K, Woodman OL, Potocnik S, Peter K, McIntyre P. Molecular Sensors of Blood Flow in Endothelial Cells. Trends Mol Med 2017; 23:850-868. [PMID: 28811171 DOI: 10.1016/j.molmed.2017.07.007] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/16/2017] [Accepted: 07/19/2017] [Indexed: 01/08/2023]
Abstract
Mechanical stress from blood flow has a significant effect on endothelial physiology, with a key role in initiating vasoregulatory signals. Disturbances in blood flow, such as in regions of disease-associated stenosis, arterial branch points, and sharp turns, can induce proatherogenic phenotypes in endothelial cells. The disruption of vascular homeostasis as a result of endothelial dysfunction may contribute to early and late stages of atherosclerosis, the underlying cause of coronary artery disease. In-depth knowledge of the mechanobiology of endothelial cells is essential to identifying mechanosensory complexes involved in the pathogenesis of atherosclerosis. In this review, we describe different blood flow patterns and summarize current knowledge on mechanosensory molecules regulating endothelial vasoregulatory functions, with clinical implications. Such information may help in the search for novel therapeutic approaches.
Collapse
Affiliation(s)
- Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia.
| | | | - Owen L Woodman
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Simon Potocnik
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Karlheinz Peter
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Peter McIntyre
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| |
Collapse
|
224
|
Abstract
Notch signaling is adjusted to different physiological contexts by expression patterns of Notch ligands and receptors, as well as by posttranslational modifications that modulate the ligand/receptor affinity. In this issue of The EMBO Journa l, Suckling et al (2017) show that an interaction of Notch ligands with membrane lipids promotes Notch binding and activation, thus proposing a new mode of Notch activity regulation.
Collapse
Affiliation(s)
- Ben‐Zion Shilo
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - David Sprinzak
- Department of Biochemistry and Molecular BiologyThe George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
225
|
Takeuchi H, Yu H, Hao H, Takeuchi M, Ito A, Li H, Haltiwanger RS. O-Glycosylation modulates the stability of epidermal growth factor-like repeats and thereby regulates Notch trafficking. J Biol Chem 2017; 292:15964-15973. [PMID: 28729422 DOI: 10.1074/jbc.m117.800102] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/18/2017] [Indexed: 11/06/2022] Open
Abstract
Glycosylation in the endoplasmic reticulum (ER) is closely associated with protein folding and quality control. We recently described a non-canonical ER quality control mechanism for folding of thrombospondin type 1 repeats by protein O-fucosyltransferase 2 (POFUT2). Epidermal growth factor-like (EGF) repeats are also small cysteine-rich protein motifs that can be O-glycosylated by several ER-localized enzymes, including protein O-glucosyltransferase 1 (POGLUT1) and POFUT1. Both POGLUT1 and POFUT1 modify the Notch receptor on multiple EGF repeats and are essential for full Notch function. The fact that POGLUT1 and POFUT1 can distinguish between folded and unfolded EGF repeats raised the possibility that they participate in a quality control pathway for folding of EGF repeats in proteins such as Notch. Here, we demonstrate that cell-surface expression of endogenous Notch1 in HEK293T cells is dependent on the presence of POGLUT1 and POFUT1 in an additive manner. In vitro unfolding assays reveal that addition of O-glucose or O-fucose stabilizes a single EGF repeat and that addition of both O-glucose and O-fucose enhances stability in an additive manner. Finally, we solved the crystal structure of a single EGF repeat covalently modified by a full O-glucose trisaccharide at 2.2 Å resolution. The structure reveals that the glycan fills up a surface groove of the EGF with multiple contacts with the protein, providing a chemical basis for the stabilizing effects of the glycans. Taken together, this work suggests that O-fucose and O-glucose glycans cooperatively stabilize individual EGF repeats through intramolecular interactions, thereby regulating Notch trafficking in cells.
Collapse
Affiliation(s)
- Hideyuki Takeuchi
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Hongjun Yu
- the Cryo-EM Structural Biology Laboratory, Van Andel Research Institute, Grand Rapids, Michigan 49503, and
| | - Huilin Hao
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Megumi Takeuchi
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Atsuko Ito
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Huilin Li
- the Cryo-EM Structural Biology Laboratory, Van Andel Research Institute, Grand Rapids, Michigan 49503, and
| | - Robert S Haltiwanger
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, .,the Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794
| |
Collapse
|
226
|
Reprogramming cellular functions with engineered membrane proteins. Curr Opin Biotechnol 2017; 47:92-101. [PMID: 28709113 DOI: 10.1016/j.copbio.2017.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/13/2017] [Indexed: 12/31/2022]
Abstract
Taking inspiration from Nature, synthetic biology utilizes and modifies biological components to expand the range of biological functions for engineering new practical devices and therapeutics. While early breakthroughs mainly concerned the design of gene circuits, recent efforts have focused on engineering signaling pathways to reprogram cellular functions. Since signal transduction across cell membranes initiates and controls intracellular signaling, membrane receptors have been targeted by diverse protein engineering approaches despite limited mechanistic understanding of their function. The modular architecture of several receptor families has enabled the empirical construction of chimeric receptors combining domains from distinct native receptors which have found successful immunotherapeutic applications. Meanwhile, progress in membrane protein structure determination, computational modeling and rational design promise to foster the engineering of a broader range of membrane receptor functions. Marrying empirical and rational membrane protein engineering approaches should enable the reprogramming of cells with widely diverse fine-tuned functions.
Collapse
|
227
|
Vieceli Dalla Sega F, Aquila G, Fortini F, Vaccarezza M, Secchiero P, Rizzo P, Campo G. Context-dependent function of ROS in the vascular endothelium: The role of the Notch pathway and shear stress. Biofactors 2017; 43:475-485. [PMID: 28419584 DOI: 10.1002/biof.1359] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/12/2017] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) act as signal molecules in several biological processes whereas excessive, unregulated, ROS production contributes to the development of pathological conditions including endothelial dysfunction and atherosclerosis. The maintenance of a healthy endothelium depends on many factors and on their reciprocal interactions; in this framework, the Notch pathway and shear stress (SS) play two lead roles. Recently, evidence of a crosstalk between ROS, Notch, and SS, is emerging. The aim of this review is to describe the way ROS interact with the Notch pathway and SS protecting from-or promoting-the development of endothelial dysfunction. © 2017 BioFactors, 43(4):475-485, 2017.
Collapse
Affiliation(s)
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Vaccarezza
- Faculty of Health Sciences, School of Biomedical Sciences, Curtin University, Perth, Australia
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA) Center, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, (RA), Italy
| | - Gianluca Campo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria S. Anna, Cona, (FE), Italy
| |
Collapse
|
228
|
Abstract
Cells differentiate into specific and functional lineages to build up tissues. It has been shown in several tissues that mitochondrial morphology, levels of "mitochondria-shaping" proteins, and mitochondrial functions change upon differentiation. In this review, we highlight the significance of mitochondrial dynamics and functions in tissue development, cell differentiation, and reprogramming processes. Signalling cascades are critical for tissue stem cell maintenance and cell fate determination, and growing evidence demonstrates mitochondria could act as a centre of intra and extracellular signals to coordinate signalling pathways, such as Notch, Wnt, and YAP/TAZ signalling. Just an organelle, however, emerges as a master regulator of cell differentiation, and can be a target to manipulate cell fates.
Collapse
Affiliation(s)
| | - Atsuko Kasahara
- Institute for Frontier Science Initiative, Cancer Research Institute, Kanazawa University, 920-1192 Kanazawa, Japan.
| |
Collapse
|
229
|
Chastagner P, Rubinstein E, Brou C. Ligand-activated Notch undergoes DTX4-mediated ubiquitylation and bilateral endocytosis before ADAM10 processing. Sci Signal 2017; 10:10/483/eaag2989. [DOI: 10.1126/scisignal.aag2989] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
230
|
Abstract
Mechanical forces play key roles in regulating cellular pathways but are challenging to study using standard biological approaches. In a recent issue of Cell, Seo et al. (2016) present a platform for in vivo single-molecule manipulation, using magnetoplasmonic nanoparticles capable of imaging, localizing, and force-loading receptor proteins at high spatiotemporal resolution.
Collapse
Affiliation(s)
- Darren Yang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Wesley P Wong
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
231
|
Abstract
Solid organ and allogeneic hematopoietic cell transplantation have become standard therapeutic interventions that save patient lives and improve quality of life. Our enhanced understanding of transplantation immunobiology has refined clinical management and improved outcomes. However, organ rejection and graft-versus-host disease remain major obstacles to the broader successful application of these therapeutic procedures. Notch signaling regulates multiple aspects of adaptive and innate immunity. Preclinical studies identified Notch signaling as a promising target in autoimmune diseases, as well as after allogeneic hematopoietic cell and solid organ transplantation. Notch was found to be a central regulator of alloreactivity across clinically relevant models of transplantation. Notch inhibition in T cells prevented graft-versus-host disease and organ rejection, establishing organ tolerance by skewing CD4 T helper polarization away from a proinflammatory response toward suppressive regulatory T cells. Notch ligand blockade also dampened alloantibody deposition and prevented chronic rejection through humoral mechanisms. Toxicities of systemic Notch blockade were observed with γ-secretase inhibitors in preclinical and early clinical trials across different indications, but they did not arise upon preclinical targeting of Delta-like Notch ligands, a strategy sufficient to confer full benefits of Notch ablation in T cell alloimmunity. Because multiple clinical grade reagents have been developed to target individual Notch ligands and receptors, the benefits of Notch blockade in transplantation are calling for translation of preclinical findings into human transplantation medicine.
Collapse
|
232
|
Lovendahl KN, Hayward AN, Gordon WR. Sequence-Directed Covalent Protein-DNA Linkages in a Single Step Using HUH-Tags. J Am Chem Soc 2017; 139:7030-7035. [PMID: 28481515 DOI: 10.1021/jacs.7b02572] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We present a robust strategy to covalently link proteins and DNA using HUH-endonuclease domains as fusion partners (HUH-tags). We show that HUH-tags react robustly with specific sequences of unmodified single-stranded DNA, and we have identified five tags that react orthogonally with distinct DNA sequences. We demonstrate the versatility of HUH-tags as fusion partners in Cas9-mediated gene editing and the construction of doubly DNA-tethered proteins for single-molecule studies. Finally we demonstrate application to cellular imaging in live and fixed cells.
Collapse
Affiliation(s)
- Klaus N Lovendahl
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota , Minneapolis, Minnesota 55455, United States
| |
Collapse
|
233
|
Ohashi K, Fujiwara S, Mizuno K. Roles of the cytoskeleton, cell adhesion and rho signalling in mechanosensing and mechanotransduction. J Biochem 2017; 161:245-254. [PMID: 28082721 DOI: 10.1093/jb/mvw082] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/13/2016] [Indexed: 11/13/2022] Open
Abstract
All cells sense and respond to various mechanical forces in and mechanical properties of their environment. To respond appropriately, cells must be able to sense the location, direction, strength and duration of these forces. Recent progress in mechanobiology has provided a better understanding of the mechanisms of mechanoresponses underlying many cellular and developmental processes. Various roles of mechanoresponses in development and tissue homeostasis have been elucidated, and many molecules involved in mechanotransduction have been identified. However, the whole picture of the functions and molecular mechanisms of mechanotransduction remains to be understood. Recently, novel mechanisms for sensing and transducing mechanical stresses via the cytoskeleton, cell-substrate and cell-cell adhesions and related proteins have been identified. In this review, we outline the roles of the cytoskeleton, cell-substrate and cell-cell adhesions, and related proteins in mechanosensing and mechanotransduction. We also describe the roles and regulation of Rho-family GTPases in mechanoresponses.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Sachiko Fujiwara
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan.,Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka 560-8531, Japan Osaka
| | - Kensaku Mizuno
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
234
|
Development of an optimized synthetic Notch receptor as an in vivo cell-cell contact sensor. Proc Natl Acad Sci U S A 2017; 114:5467-5472. [PMID: 28490499 DOI: 10.1073/pnas.1703205114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Detection and manipulation of direct cell-cell contact in complex tissues is a fundamental and challenging problem in many biological studies. Here, we report an optimized Notch-based synthetic receptor (synNQ) useful to study direct cell-cell interactions in Drosophila With the synNQ system, cells expressing a synthetic receptor, which contains Notch activation machinery and a downstream transcriptional activator, QF, are activated by a synthetic GFP ligand expressed by contacting neighbor cells. To avoid cis-inhibition, mutually exclusive expression of the synthetic ligand and receptor is achieved using the "flippase-out" system. Expression of the synthetic GFP ligand is controlled by the Gal4/UAS system for easy and broad applications. Using synNQ, we successfully visualized cell-cell interactions within and between most fly tissues, revealing previously undocumented cell-cell contacts. Importantly, in addition to detection of cells in contact with one another, synNQ allows for genetic manipulation in all cells in contact with a targeted cell population, which we demonstrate in the context of cell competition in developing wing disks. Altogether, the synNQ genetic system will enable a broad range of studies of cell contact in developmental biology.
Collapse
|
235
|
Chen KY, Srinivasan T, Tung KL, Belmonte JM, Wang L, Murthy PKL, Choi J, Rakhilin N, King S, Varanko AK, Witherspoon M, Nishimura N, Glazier JA, Lipkin SM, Bu P, Shen X. A Notch positive feedback in the intestinal stem cell niche is essential for stem cell self-renewal. Mol Syst Biol 2017; 13:927. [PMID: 28455349 PMCID: PMC5408779 DOI: 10.15252/msb.20167324] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 01/14/2023] Open
Abstract
The intestinal epithelium is the fastest regenerative tissue in the body, fueled by fast-cycling stem cells. The number and identity of these dividing and migrating stem cells are maintained by a mosaic pattern at the base of the crypt. How the underlying regulatory scheme manages this dynamic stem cell niche is not entirely clear. We stimulated intestinal organoids with Notch ligands and inhibitors and discovered that intestinal stem cells employ a positive feedback mechanism via direct Notch binding to the second intron of the Notch1 gene. Inactivation of the positive feedback by CRISPR/Cas9 mutation of the binding sequence alters the mosaic stem cell niche pattern and hinders regeneration in organoids. Dynamical system analysis and agent-based multiscale stochastic modeling suggest that the positive feedback enhances the robustness of Notch-mediated niche patterning. This study highlights the importance of feedback mechanisms in spatiotemporal control of the stem cell niche.
Collapse
Affiliation(s)
- Kai-Yuan Chen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tara Srinivasan
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Kuei-Ling Tung
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Julio M Belmonte
- Biocomplexity Institute and Department of Physics, Indiana University, Bloomington, IN, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | | | - Jiahn Choi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikolai Rakhilin
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sarah King
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Mavee Witherspoon
- School of Mechanical Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Nozomi Nishimura
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - James A Glazier
- Biocomplexity Institute and Department of Physics, Indiana University, Bloomington, IN, USA
| | - Steven M Lipkin
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Pengcheng Bu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiling Shen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
236
|
Notch and Hippo signaling converge on Strawberry Notch 1 (Sbno1) to synergistically activate Cdx2 during specification of the trophectoderm. Sci Rep 2017; 7:46135. [PMID: 28401892 PMCID: PMC5389439 DOI: 10.1038/srep46135] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 03/13/2017] [Indexed: 02/08/2023] Open
Abstract
The first binary cell fate decision occurs at the morula stage and gives rise to two distinct types of cells that constitute the trophectoderm (TE) and inner cell mass (ICM). The cell fate determinant, Cdx2, is induced in TE cells and plays an essential role in their differentiation and maintenance. Notch and Hippo signaling cascades are assumed to converge onto regulatory elements of Cdx2, however, the underlying molecular mechanisms are largely unknown. Here, we show involvement of Strawberry Notch1 (Sbno1), a novel chromatin factor of the helicase superfamily 2, during preimplantation development. Sbno1 knockout embryos die at the preimplantation stage without forming a blastocoel, and Cdx2 is not turned on even though both Yap and Tead4 reside normally in nuclei. Accordingly, Sbno1 acts on the trophectoderm-enhancer (TEE) of Cdx2, ensuring its robust and synergistic activation by the Yap/Tead4 and NICD/Rbpj complexes. Interestingly, this synergism is enhanced when cells are mechanically stretched, which might reflect that TE cells are continuously stretched by the expanding ICM and blastocoel cavity. In addition, the histone chaperone, FACT (FAcilitates Chromatin Transcription) physically interacts with Sbno1. Our data provide new evidence on TE specification, highlighting unexpected but essential functions of the highly conserved chromatin factor, Sbno1.
Collapse
|
237
|
Millon-Frémillon A, Aureille J, Guilluy C. Analyzing Cell Surface Adhesion Remodeling in Response to Mechanical Tension Using Magnetic Beads. J Vis Exp 2017:55330. [PMID: 28362397 PMCID: PMC5408950 DOI: 10.3791/55330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mechanosensitive cell surface adhesion complexes allow cells to sense the mechanical properties of their surroundings. Recent studies have identified both force-sensing molecules at adhesion sites, and force-dependent transcription factors that regulate lineage-specific gene expression and drive phenotypic outputs. However, the signaling networks converting mechanical tension into biochemical pathways have remained elusive. To explore the signaling pathways engaged upon mechanical tension applied to cell surface receptor, superparamagnetic microbeads can be used. Here we present a protocol for using magnetic beads to apply forces to cell surface adhesion proteins. Using this approach, it is possible to investigate not only force-dependent cytoplasmic signaling pathways by various biochemical approaches, but also adhesion remodeling by magnetic isolation of adhesion complexes attached to the ligand-coated beads. This protocol includes the preparation of ligand-coated superparamagnetic beads, and the application of define tensile forces followed by biochemical analyses. Additionally, we provide a representative sample of data demonstrating that tension applied to integrin-based adhesion triggers adhesion remodeling and alters protein tyrosine phosphorylation.
Collapse
Affiliation(s)
| | - Julien Aureille
- Institute for Advanced Biosciences, Centre de recherche UGA - INSERM U1209 - CNRS UMR
| | - Christophe Guilluy
- Institute for Advanced Biosciences, Centre de recherche UGA - INSERM U1209 - CNRS UMR;
| |
Collapse
|
238
|
Sadelain M. Chimeric Antigen Receptors: A Paradigm Shift in Immunotherapy. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-050216-034351] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
239
|
Luca VC, Kim BC, Ge C, Kakuda S, Wu D, Roein-Peikar M, Haltiwanger RS, Zhu C, Ha T, Garcia KC. Notch-Jagged complex structure implicates a catch bond in tuning ligand sensitivity. Science 2017; 355:1320-1324. [PMID: 28254785 DOI: 10.1126/science.aaf9739] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 02/17/2017] [Indexed: 12/18/2022]
Abstract
Notch receptor activation initiates cell fate decisions and is distinctive in its reliance on mechanical force and protein glycosylation. The 2.5-angstrom-resolution crystal structure of the extracellular interacting region of Notch1 complexed with an engineered, high-affinity variant of Jagged1 (Jag1) reveals a binding interface that extends ~120 angstroms along five consecutive domains of each protein. O-Linked fucose modifications on Notch1 epidermal growth factor-like (EGF) domains 8 and 12 engage the EGF3 and C2 domains of Jag1, respectively, and different Notch1 domains are favored in binding to Jag1 than those that bind to the Delta-like 4 ligand. Jag1 undergoes conformational changes upon Notch binding, exhibiting catch bond behavior that prolongs interactions in the range of forces required for Notch activation. This mechanism enables cellular forces to regulate binding, discriminate among Notch ligands, and potentiate Notch signaling.
Collapse
Affiliation(s)
- Vincent C Luca
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Byoung Choul Kim
- Howard Hughes Medical Institute, Baltimore, MD 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chenghao Ge
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shinako Kakuda
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Di Wu
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Mehdi Roein-Peikar
- Howard Hughes Medical Institute, Baltimore, MD 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Cheng Zhu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Taekjip Ha
- Howard Hughes Medical Institute, Baltimore, MD 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Howard Hughes Medical Institute, Stanford, CA 94305, USA
| |
Collapse
|
240
|
Lee D, Huang TH, De La Cruz A, Callejas A, Lois C. Methods to investigate the structure and connectivity of the nervous system. Fly (Austin) 2017; 11:224-238. [PMID: 28277925 PMCID: PMC5552278 DOI: 10.1080/19336934.2017.1295189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Understanding the computations that take place in neural circuits requires identifying how neurons in those circuits are connected to one another. In addition, recent research indicates that aberrant neuronal wiring may be the cause of several neurodevelopmental disorders, further emphasizing the importance of identifying the wiring diagrams of brain circuits. To address this issue, several new approaches have been recently developed. In this review, we describe several methods that are currently available to investigate the structure and connectivity of the brain, and discuss their strengths and limitations.
Collapse
Affiliation(s)
- Donghyung Lee
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Ting-Hao Huang
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Aubrie De La Cruz
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Antuca Callejas
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA.,b Department of Cell Biology, School of Science , University of Extremadura , Badajoz , Spain
| | - Carlos Lois
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| |
Collapse
|
241
|
Aster JC, Pear WS, Blacklow SC. The Varied Roles of Notch in Cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:245-275. [PMID: 27959635 DOI: 10.1146/annurev-pathol-052016-100127] [Citation(s) in RCA: 514] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Notch receptors influence cellular behavior by participating in a seemingly simple signaling pathway, but outcomes produced by Notch signaling are remarkably varied depending on signal dose and cell context. Here, after briefly reviewing new insights into physiologic mechanisms of Notch signaling in healthy tissues and defects in Notch signaling that contribute to congenital disorders and viral infection, we discuss the varied roles of Notch in cancer, focusing on cell autonomous activities that may be either oncogenic or tumor suppressive.
Collapse
Affiliation(s)
- Jon C Aster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115;
| | - 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 19104
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
242
|
Hadjivasiliou Z, Hunter GL, Baum B. A new mechanism for spatial pattern formation via lateral and protrusion-mediated lateral signalling. J R Soc Interface 2016; 13:20160484. [PMID: 27807273 PMCID: PMC5134009 DOI: 10.1098/rsif.2016.0484] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023] Open
Abstract
Tissue organization and patterning are critical during development when genetically identical cells take on different fates. Lateral signalling plays an important role in this process by helping to generate self-organized spatial patterns in an otherwise uniform collection of cells. Recent data suggest that lateral signalling can be mediated both by junctional contacts between neighbouring cells and via cellular protrusions that allow non-neighbouring cells to interact with one another at a distance. However, it remains unclear precisely how signalling mediated by these distinct types of cell-cell contact can physically contribute to the generation of complex patterns without the assistance of diffusible morphogens or pre-patterns. To explore this question, in this work we develop a model of lateral signalling based on a single receptor/ligand pair as exemplified by Notch and Delta. We show that allowing the signalling kinetics to differ at junctional versus protrusion-mediated contacts, an assumption inspired by recent data which show that the cleavage of Notch in several systems requires both Delta binding and the application of mechanical force, permits individual cells to act to promote both lateral activation and lateral inhibition. Strikingly, under this model, in which Delta can sequester Notch, a variety of patterns resembling those typical of reaction-diffusion systems is observed, together with more unusual patterns that arise when we consider changes in signalling kinetics, and in the length and distribution of protrusions. Importantly, these patterns are self-organizing-so that local interactions drive tissue-scale patterning. Together, these data show that protrusions can, in principle, generate different types of patterns in addition to contributing to long-range signalling and to pattern refinement.
Collapse
Affiliation(s)
- Zena Hadjivasiliou
- Centre for Mathematics, Physics, and Engineering in the Life Sciences and Experimental Biology, University College London, London WC1E 6BT, UK
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - Ginger L Hunter
- MRC-Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
- Institute of Physics of Living Systems, University College London, London WC1E 6BT, UK
| | - Buzz Baum
- MRC-Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
- Institute of Physics of Living Systems, University College London, London WC1E 6BT, UK
| |
Collapse
|
243
|
LaFoya B, Munroe JA, Mia MM, Detweiler MA, Crow JJ, Wood T, Roth S, Sharma B, Albig AR. Notch: A multi-functional integrating system of microenvironmental signals. Dev Biol 2016; 418:227-41. [PMID: 27565024 PMCID: PMC5144577 DOI: 10.1016/j.ydbio.2016.08.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/15/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022]
Abstract
The Notch signaling cascade is an evolutionarily ancient system that allows cells to interact with their microenvironmental neighbors through direct cell-cell interactions, thereby directing a variety of developmental processes. Recent research is discovering that Notch signaling is also responsive to a broad variety of stimuli beyond cell-cell interactions, including: ECM composition, crosstalk with other signaling systems, shear stress, hypoxia, and hyperglycemia. Given this emerging understanding of Notch responsiveness to microenvironmental conditions, it appears that the classical view of Notch as a mechanism enabling cell-cell interactions, is only a part of a broader function to integrate microenvironmental cues. In this review, we summarize and discuss published data supporting the idea that the full function of Notch signaling is to serve as an integrator of microenvironmental signals thus allowing cells to sense and respond to a multitude of conditions around them.
Collapse
Affiliation(s)
- Bryce LaFoya
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Jordan A Munroe
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Masum M Mia
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Michael A Detweiler
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Jacob J Crow
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Travis Wood
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Steven Roth
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Bikram Sharma
- Department of Biological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Allan R Albig
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA; Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|
244
|
Roybal KT, Williams JZ, Morsut L, Rupp LJ, Kolinko I, Choe JH, Walker WJ, McNally KA, Lim WA. Engineering T Cells with Customized Therapeutic Response Programs Using Synthetic Notch Receptors. Cell 2016; 167:419-432.e16. [PMID: 27693353 DOI: 10.1016/j.cell.2016.09.011] [Citation(s) in RCA: 509] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 01/17/2023]
Abstract
Redirecting T cells to attack cancer using engineered chimeric receptors provides powerful new therapeutic capabilities. However, the effectiveness of therapeutic T cells is constrained by the endogenous T cell response: certain facets of natural response programs can be toxic, whereas other responses, such as the ability to overcome tumor immunosuppression, are absent. Thus, the efficacy and safety of therapeutic cells could be improved if we could custom sculpt immune cell responses. Synthetic Notch (synNotch) receptors induce transcriptional activation in response to recognition of user-specified antigens. We show that synNotch receptors can be used to sculpt custom response programs in primary T cells: they can drive a la carte cytokine secretion profiles, biased T cell differentiation, and local delivery of non-native therapeutic payloads, such as antibodies, in response to antigen. SynNotch T cells can thus be used as a general platform to recognize and remodel local microenvironments associated with diverse diseases.
Collapse
Affiliation(s)
- Kole T Roybal
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Jasper Z Williams
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Leonardo Morsut
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Levi J Rupp
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Isabel Kolinko
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Joseph H Choe
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Whitney J Walker
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Krista A McNally
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Wendell A Lim
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; UCSF Center for Systems and Synthetic Biology, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, San Francisco, CA 94158, USA.
| |
Collapse
|
245
|
Huang TH, Velho T, Lois C. Monitoring cell-cell contacts in vivo in transgenic animals. Development 2016; 143:4073-4084. [PMID: 27660327 DOI: 10.1242/dev.142406] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/13/2016] [Indexed: 12/15/2022]
Abstract
We used a synthetic genetic system based on ligand-induced intramembrane proteolysis to monitor cell-cell contacts in animals. Upon ligand-receptor interaction in sites of cell-cell contact, the transmembrane domain of an engineered receptor is cleaved by intramembrane proteolysis and releases a protein fragment that regulates transcription in the interacting partners. We demonstrate that the system can be used to regulate gene expression between interacting cells, both in vitro and in vivo, in transgenic Drosophila We show that the system allows for detection of interactions between neurons and glia in the Drosophila nervous system. In addition, we observed that when the ligand is expressed in subsets of neurons with a restricted localization in the brain it leads to activation of transcription in a selected set of glial cells that interact with those neurons. This system will be useful to monitor cell-cell interactions in animals, and can be used to genetically manipulate cells that interact with one another.
Collapse
Affiliation(s)
- Ting-Hao Huang
- California Institute of Technology, Division of Biology and Biological Engineering, Beckman Institute MC 139-74, 1200 East California Blvd, Pasadena, CA 91125, USA.,Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Tarciso Velho
- California Institute of Technology, Division of Biology and Biological Engineering, Beckman Institute MC 139-74, 1200 East California Blvd, Pasadena, CA 91125, USA.,Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59056-450, Brazil
| | - Carlos Lois
- California Institute of Technology, Division of Biology and Biological Engineering, Beckman Institute MC 139-74, 1200 East California Blvd, Pasadena, CA 91125, USA
| |
Collapse
|
246
|
Esteves de Lima J, Bonnin MA, Birchmeier C, Duprez D. Muscle contraction is required to maintain the pool of muscle progenitors via YAP and NOTCH during fetal myogenesis. eLife 2016; 5. [PMID: 27554485 PMCID: PMC5030091 DOI: 10.7554/elife.15593] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/23/2016] [Indexed: 12/27/2022] Open
Abstract
The importance of mechanical activity in the regulation of muscle progenitors during chick development has not been investigated. We show that immobilization decreases NOTCH activity and mimics a NOTCH loss-of-function phenotype, a reduction in the number of muscle progenitors and increased differentiation. Ligand-induced NOTCH activation prevents the reduction of muscle progenitors and the increase of differentiation upon immobilization. Inhibition of NOTCH ligand activity in muscle fibers suffices to reduce the progenitor pool. Furthermore, immobilization reduces the activity of the transcriptional co-activator YAP and the expression of the NOTCH ligand JAG2 in muscle fibers. YAP forced-activity in muscle fibers prevents the decrease of JAG2 expression and the number of PAX7+ cells in immobilization conditions. Our results identify a novel mechanism acting downstream of muscle contraction, where YAP activates JAG2 expression in muscle fibers, which in turn regulates the pool of fetal muscle progenitors via NOTCH in a non-cell-autonomous manner. DOI:http://dx.doi.org/10.7554/eLife.15593.001 Skeletal muscle is attached to the skeleton and allows the body to move. Making a new muscle, or repairing an existing one, relies on stem cells that are present inside muscles. A major goal of skeletal muscle research is to understand the signals that regulate the abilities of muscle stem cells to divide and give rise to more stem cells or to become muscle cells. Molecular signals are known to regulate the numbers of stem cells in the muscle. Skeletal muscles become larger if they are exercised, but it is not clear if mechanical forces generated by muscle contractions directly affect the number of muscle stem cells. The NOTCH signaling pathway contributes to maintaining the population of stem cells in muscles by forcing the stem cells to divide and preventing them from becoming muscle cells. Here, Esteves de Lima et al. investigated whether muscle contraction regulates NOTCH signaling during muscle formation in chick fetuses. The experiments show that muscle contraction stimulates the activity of a protein called YAP in muscle cells, which in turn, activates a gene in the NOTCH signaling pathway known as JAG2. This increases NOTCH signaling activity in the neighboring stem cells and maintains the number of stem cells in the muscle. The next step following this work will be to establish if this mechanism also operates during muscle formation and regeneration in other animals such as mice and zebrafish. DOI:http://dx.doi.org/10.7554/eLife.15593.002
Collapse
Affiliation(s)
- Joana Esteves de Lima
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| | - Marie-Ange Bonnin
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| | - Carmen Birchmeier
- Developmental Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Delphine Duprez
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| |
Collapse
|
247
|
Abstract
The highly conserved Notch signalling pathway functions in many different developmental and homeostatic processes, which raises the question of how this pathway can achieve such diverse outcomes. With a direct route from the membrane to the nucleus, the Notch pathway has fewer opportunities for regulation than do many other signalling pathways, yet it generates exquisitely patterned structures, including sensory hair cells and branched arterial networks. More confusingly, its activity promotes tissue growth and cancers in some circumstances but cell death and tumour suppression in others. Many different regulatory mechanisms help to shape the activity of the Notch pathway, generating functional outputs that are appropriate for each context. These mechanisms include the receptor-ligand landscape, the tissue topology, the nuclear environment and the connectivity of the regulatory networks.
Collapse
Affiliation(s)
- Sarah J Bray
- Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
248
|
Abstract
The Notch signaling pathway plays fundamental roles in diverse developmental processes. Studies of the basic biology of Notch function have provided insights into how its dysfunction contributes to multi-systemic diseases and cancer. In addition, our understanding of Notch signaling in maintaining stem/progenitor cell populations is revealing new avenues for rekindling regeneration. The Notch IX meeting, which was held in Athens, Greece in October 2015, brought together scientists working on different model systems and studying Notch signaling in various contexts. Here, we provide a summary of the key points that were presented at the meeting. Although we focus on the molecular mechanisms that determine Notch signaling and its role in development, we also cover talks describing roles for Notch in adulthood. Together, the talks revealed how interactions between adjacent cells mediated by Notch regulate development and physiology at multiple levels.
Collapse
Affiliation(s)
- Ajay Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Building 6B, Room 3B-315, Bethesda, MD 20892, USA
| | - Laure Balle-Cuif
- Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS - Université Paris-Sud, Avenue de la Terrasse, Bldg 5, 91190 Gif-sur-Yvette, France
| |
Collapse
|
249
|
Mrkonjic S, Destaing O, Albiges-Rizo C. Mechanotransduction pulls the strings of matrix degradation at invadosome. Matrix Biol 2016; 57-58:190-203. [PMID: 27392543 DOI: 10.1016/j.matbio.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023]
Abstract
Degradation of the extracellular matrix is a critical step of tumor cell invasion. Both protease-dependent and -independent mechanisms have been described as alternate processes in cancer cell motility. Interestingly, some effectors of protease-dependent degradation are focalized at invadosomes and are directly coupled with contractile and adhesive machineries composed of multiple mechanosensitive proteins. This review presents recent findings in protease-dependent mechanisms elucidating the ways the force affects extracellular matrix degradation by targeting protease expression and activity at invadosome. The aim is to highlight mechanosensing and mechanotransduction processes to direct the degradative activity at invadosomes, with the focus on membrane tension, proteases and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Sanela Mrkonjic
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France
| | - Olivier Destaing
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| | - Corinne Albiges-Rizo
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| |
Collapse
|
250
|
Sadelain M. Chimeric antigen receptors: driving immunology towards synthetic biology. Curr Opin Immunol 2016; 41:68-76. [PMID: 27372731 DOI: 10.1016/j.coi.2016.06.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/15/2022]
Abstract
The advent of second generation chimeric antigen receptors and the CD19 paradigm have ushered a new therapeutic modality in oncology. In contrast to earlier forms of adoptive cell therapy, which were based on the isolation and expansion of naturally occurring T cells, CAR therapy is based on the design and manufacture of engineered T cells with optimized properties. A new armamentarium, comprising not only CARs but also chimeric costimulatory receptors, chimeric cytokine receptors, inhibitory receptors and synthetic Notch receptors, expressed in naïve, central memory or stem cell-like memory T cells, is being developed for clinical use in a wide range of cancers. Immunological principles are thus finding a new purpose thanks to advances in genetic engineering, synthetic biology and cell manufacturing sciences.
Collapse
Affiliation(s)
- Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|