1
|
Hill AJ, Robinson B, Jones JG, Sternberg PW, Van Buskirk C. Sleep drive is coupled to tissue damage via shedding of Caenorhabditis elegans EGFR ligand SISS-1. Nat Commun 2024; 15:10886. [PMID: 39738055 PMCID: PMC11686035 DOI: 10.1038/s41467-024-55252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/04/2024] [Indexed: 01/01/2025] Open
Abstract
The benefits of sleep extend beyond the nervous system. Peripheral tissues impact sleep regulation, and increased sleep is observed in response to damaging conditions, even those that selectively affect non-neuronal cells. However, the 'sleep need' signal released by stressed tissues is not known. Sleep in the nematode C. elegans is independent of circadian cues and can be triggered rapidly by damaging conditions. This stress-induced sleep is mediated by neurons that require the Epidermal Growth Factor Receptor (EGFR) for their sleep-promoting function, but the only known C. elegans EGFR ligand, LIN-3, is not required for sleep. Here we describe SISS-1 (stress-induced sleepless), an EGF family ligand that is required for stress-induced sleep. We show that SISS-1 overexpression induces sleep in an EGFR-dependent, sleep neuron-dependent manner. We find that SISS-1 undergoes stress-responsive shedding by the ADM-4/ADAM17 metalloprotease, and that the ADM-4 site of action depends on the tissue specificity of the stressor. Our findings support a model in which SISS-1 is released from damaged tissues to activate EGFR in sleep neurons, identifying a molecular link between cellular stress and organismal sleep drive. Our data also point to a mechanism insulating this sleep signal from EGFR-mediated signaling during development.
Collapse
Affiliation(s)
- Andrew J Hill
- Department of Biology, California State University Northridge, Northridge, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Bryan Robinson
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Jesse G Jones
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Cheryl Van Buskirk
- Department of Biology, California State University Northridge, Northridge, CA, USA.
| |
Collapse
|
2
|
Brinkley DM, Smith KC, Fink EC, Kwen W, Yoo NH, West Z, Sullivan NL, Farthing AS, Hale VA, Goutte C. Notch signaling without the APH-2/nicastrin subunit of gamma secretase in Caenorhabditis elegans germline stem cells. Genetics 2024; 227:iyae076. [PMID: 38717968 PMCID: PMC12098933 DOI: 10.1093/genetics/iyae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/01/2024] [Indexed: 07/09/2024] Open
Abstract
The final step in Notch signaling activation is the transmembrane cleavage of Notch receptor by γ secretase. Thus far, genetic and biochemical evidence indicates that four subunits are essential for γ secretase activity in vivo: presenilin (the catalytic core), APH-1, PEN-2, and APH-2/nicastrin. Although some γ secretase activity has been detected in APH-2/nicastrin-deficient mammalian cell lines, the lack of biological relevance for this activity has left the quaternary γ secretase model unchallenged. Here, we provide the first example of in vivo Notch signal transduction without APH-2/nicastrin. The surprising dispensability of APH-2/nicastrin is observed in Caenorhabditis elegans germline stem cells (GSCs) and contrasts with its essential role in previously described C. elegans Notch signaling events. Depletion of GLP-1/Notch, presenilin, APH-1, or PEN-2 causes a striking loss of GSCs. In contrast, aph-2/nicastrin mutants maintain GSCs and exhibit robust and localized expression of the downstream Notch target sygl-1. Interestingly, APH-2/nicastrin is normally expressed in GSCs and becomes essential under conditions of compromised Notch function. Further insight is provided by reconstituting the C. elegans γ secretase complex in yeast, where we find that APH-2/nicastrin increases but is not essential for γ secretase activity. Together, our results are most consistent with a revised model of γ secretase in which the APH-2/nicastrin subunit has a modulatory, rather than obligatory role. We propose that a trimeric presenilin-APH-1-PEN-2 γ secretase complex can provide a low level of γ secretase activity, and that cellular context determines whether or not APH-2/nicastrin is essential for effective Notch signal transduction.
Collapse
Affiliation(s)
- David M Brinkley
- Program in Biochemistry and Biophysics, Amherst College, Amherst, MA 01002, USA
| | - Karen C Smith
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Emma C Fink
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Woohyun Kwen
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Nina H Yoo
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Zachary West
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Nora L Sullivan
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Alex S Farthing
- Program in Biochemistry and Biophysics, Amherst College, Amherst, MA 01002, USA
| | - Valerie A Hale
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Caroline Goutte
- Program in Biochemistry and Biophysics, Amherst College, Amherst, MA 01002, USA
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| |
Collapse
|
3
|
Gowripriya T, Meharaj Afrin K, Paurna M, Yashwanth R, Bhaskar JP, Suresh R, Balamurugan K. Regulation of miR-61 and col-19 via TGF-β and Notch signalling in Caenorhabditis elegans against Klebsiella aerogenes infection. Microb Pathog 2024; 186:106505. [PMID: 38122874 DOI: 10.1016/j.micpath.2023.106505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Klebsiella aerogenes, previously known as Enterobacter aerogenes, is a gram-negative bacterium typically present in the gastrointestinal tract. While numerous studies reported the pathogenicity and drug resistance of this bacterium there remains a lack of comprehensive research on K. aerogenes induced alterations in the host cellular mechanisms. In this study, we identify a previously uncharacterized C. elegans miR-61 that defines an evolutionarily conserved miRNA important for development and innate immunity regulation through Notch and TGF-β signaling pathway. We employed C. elegans wild-type (N2) as well as mutant strains, such as TGF-β (sma-6) and notch-signaling pathway mutants (adm-4 and mir-61). Our results have demonstrated that the K. aerogenes infected mutants exhibited significantly reduced survival rate, reduced pharyngeal pumping, altered swimming and chemotactic behavior. Moreover, K. aerogenes affects the healthspan by increasing ROS level in the mutants. The gene expression analysis revealed that K. aerogenes upregulated egl-30, tph-1 and sod-1 in adm-4, mir-61 mutants not in sma-6. The in-silico analysis indicated an interaction between mir-61 and col-19, which was confirmed by the upregulation of miR-61 expression and the downregulation of col-19 in sma-6, adm-4, and wild-type strains. These findings suggest that C. elegans activates mir-61 and col-19 regulation through the Notch and TGF-β signaling pathway against K. aerogenes infection.
Collapse
Affiliation(s)
- Thirumugam Gowripriya
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, India.
| | | | - Manikandan Paurna
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, India.
| | - Radhakrishnan Yashwanth
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | - James Prabhanand Bhaskar
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | - Ramamurthi Suresh
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | | |
Collapse
|
4
|
Rani N, Alam MM, Jamal A, Bin Ghaffar U, Parvez S. Caenorhabditis elegans: A transgenic model for studying age-associated neurodegenerative diseases. Ageing Res Rev 2023; 91:102036. [PMID: 37598759 DOI: 10.1016/j.arr.2023.102036] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Neurodegenerative diseases (NDs) are a heterogeneous group of aging-associated ailments characterized by interrupting cellular proteostasic machinery and the misfolding of distinct proteins to form toxic aggregates in neurons. Neurodegenerative diseases, which include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and others, are becoming an increasing threat to human health worldwide. The degeneration and death of certain specific groups of neurons are the hallmarks of these diseases. Over the past decades, Caenorhabditis eleganshas beenwidely used as a transgenic model to investigate biological processes related to health and disease. The nematode Caenorhabditis elegans (C. elegans) has developed as a powerful tool for studying disease mechanisms due to its ease of genetic handling and instant cultivation while providing a whole-animal system amendable to several molecular and biochemical techniques. In this review, we elucidate the potential of C. elegans as a versatile platform for systematic dissection of the molecular basis of human disease, focusing on neurodegenerative disorders, and may help better our understanding of the disease mechanisms and search for new therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Usama Bin Ghaffar
- Department of Basic Science, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
5
|
Herd CS, Yu X, Cui Y, Franz AWE. Identification of the extracellular metallo-endopeptidases ADAM and ADAMTS in the yellow fever mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 148:103815. [PMID: 35932972 PMCID: PMC11149919 DOI: 10.1016/j.ibmb.2022.103815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 06/15/2023]
Abstract
The mosquito Aedes aegypti is a major vector for dengue, Zika, yellow fever, and chikungunya (CHIKV) viruses, which cause significant morbidity and mortality among human populations in the tropical regions of the world. Following ingestion of a viremic bloodmeal from a vertebrate host, an arbovirus needs to productively infect the midgut epithelium of the mosquito. De novo synthesized virions then exit the midgut by traversing the surrounding basal lamina (BL) in order to disseminate to secondary tissues and infect those. Once the salivary glands are infected, the virus is transmitted to a vertebrate host along with saliva released during probing of the mosquito. Midgut tissue distention due to bloodmeal ingestion leads to remodeling of the midgut structure and facilitates virus dissemination from the organ. Previously, we described the matrix-metalloproteinases (MMP) of Ae. aegypti as zinc ion dependent endopeptidases (Metzincins) and showed MMP activity during midgut BL rearrangement as a consequence of bloodmeal ingestion and subsequent digestion thereby affecting arbovirus dissemination from the midgut. Here we investigate the ADAM/ADAMTS of Ae. aegypti, which form another major group of multi-domain proteinases within the Metzincin superfamily and are active during extra-cellular matrix (ECM) remodeling. Seven different ADAM and five ADAMTS were identified in Ae. aegypti. The functional protein domain structures of the identified mosquito ADAM resembled those of human ADAM10, ADAM12, and ADAM17, while two of the five mosquito ADAMTS had human orthologs. Expression profiling of Ae. aegypti ADAM/ADAMTS in immature forms, whole body-females, midguts, and ovarian tissues showed transcriptional activity of the proteinases during metamorphosis, bloodmeal ingestion/digestion, and female reproduction. Custom-made antibodies to ADAM10a and ADAM12c showed that both were strongly expressed in midgut and ovarian tissues. Furthermore, transient silencing of ADAM12c significantly reduced the carcass infection rate with CHIKV at 24 h post-infection, while silencing of ADAM12a significantly increased viral titers in secondary tissues at the same time point. Our results indicate a functional specificity for several ADAM/ADAMTS in those selected mosquito tissues.
Collapse
Affiliation(s)
- Christie S Herd
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| | - Xiudao Yu
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| | - Yingjun Cui
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| | - Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
6
|
Cho Y, Bae HG, Okun E, Arumugam TV, Jo DG. Physiology and pharmacology of amyloid precursor protein. Pharmacol Ther 2022; 235:108122. [PMID: 35114285 DOI: 10.1016/j.pharmthera.2022.108122] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
Amyloid precursor protein (APP) is an evolutionarily conserved transmembrane protein and a well-characterized precursor protein of amyloid-beta (Aβ) peptides, which accumulate in the brains of individuals with Alzheimer's disease (AD)-related pathologies. Aβ has been extensively investigated since the amyloid hypothesis in AD was proposed. Besides Aβ, previous studies on APP and its proteolytic cleavage products have suggested their diverse pathological and physiological functions. However, their roles still have not been thoroughly understood. In this review, we extensively discuss the evolutionarily-conserved biology of APP, including its structure and processing pathway, as well as recent findings on the physiological roles of APP and its fragments in the central nervous system and peripheral nervous system. We have also elaborated upon the current status of APP-targeted therapeutic approaches for AD treatment by discussing inhibitors of several proteases participating in APP processing, including α-, β-, and γ-secretases. Finally, we have highlighted the future perspectives pertaining to further research and the potential clinical role of APP.
Collapse
Affiliation(s)
- Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Han-Gyu Bae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Pauld Feder Laboratory on Alzheimer's Disease Research, Israel
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea; Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
7
|
Kiyozumi D, Ikawa M. Proteolysis in Reproduction: Lessons From Gene-Modified Organism Studies. Front Endocrinol (Lausanne) 2022; 13:876370. [PMID: 35600599 PMCID: PMC9114714 DOI: 10.3389/fendo.2022.876370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022] Open
Abstract
The physiological roles of proteolysis are not limited to degrading unnecessary proteins. Proteolysis plays pivotal roles in various biological processes through cleaving peptide bonds to activate and inactivate proteins including enzymes, transcription factors, and receptors. As a wide range of cellular processes is regulated by proteolysis, abnormalities or dysregulation of such proteolytic processes therefore often cause diseases. Recent genetic studies have clarified the inclusion of proteases and protease inhibitors in various reproductive processes such as development of gonads, generation and activation of gametes, and physical interaction between gametes in various species including yeast, animals, and plants. Such studies not only clarify proteolysis-related factors but the biological processes regulated by proteolysis for successful reproduction. Here the physiological roles of proteases and proteolysis in reproduction will be reviewed based on findings using gene-modified organisms.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
8
|
Notch3 signalling and vascular remodelling in pulmonary arterial hypertension. Clin Sci (Lond) 2020; 133:2481-2498. [PMID: 31868216 PMCID: PMC6928565 DOI: 10.1042/cs20190835] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/27/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Notch signalling is critically involved in vascular morphogenesis and function. Four Notch isoforms (Notch1–4) regulating diverse cellular processes have been identified. Of these, Notch3 is expressed almost exclusively in vascular smooth muscle cells (VSMCs), where it is critically involved in vascular development and differentiation. Under pathological conditions, Notch3 regulates VSMC switching between the contractile and synthetic phenotypes. Abnormal Notch3 signalling plays an important role in vascular remodelling, a hallmark of several cardiovascular diseases, including pulmonary arterial hypertension (PAH). Because of the importance of Notch3 in VSMC (de)differentiation, Notch3 has been implicated in the pathophysiology of pulmonary vascular remodelling in PAH. Here we review the current literature on the role of Notch in VSMC function with a focus on Notch3 signalling in pulmonary artery VSMCs, and discuss potential implications in pulmonary artery remodelling in PAH.
Collapse
|
9
|
Hubbard EJA, Schedl T. Biology of the Caenorhabditis elegans Germline Stem Cell System. Genetics 2019; 213:1145-1188. [PMID: 31796552 PMCID: PMC6893382 DOI: 10.1534/genetics.119.300238] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Stem cell systems regulate tissue development and maintenance. The germline stem cell system is essential for animal reproduction, controlling both the timing and number of progeny through its influence on gamete production. In this review, we first draw general comparisons to stem cell systems in other organisms, and then present our current understanding of the germline stem cell system in Caenorhabditis elegans In contrast to stereotypic somatic development and cell number stasis of adult somatic cells in C. elegans, the germline stem cell system has a variable division pattern, and the system differs between larval development, early adult peak reproduction and age-related decline. We discuss the cell and developmental biology of the stem cell system and the Notch regulated genetic network that controls the key decision between the stem cell fate and meiotic development, as it occurs under optimal laboratory conditions in adult and larval stages. We then discuss alterations of the stem cell system in response to environmental perturbations and aging. A recurring distinction is between processes that control stem cell fate and those that control cell cycle regulation. C. elegans is a powerful model for understanding germline stem cells and stem cell biology.
Collapse
Affiliation(s)
- E Jane Albert Hubbard
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology and Pathology, New York University School of Medicine, New York 10016
| | - Tim Schedl
- and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
10
|
Reinhardt S, Schuck F, Stoye N, Hartmann T, Grimm MOW, Pflugfelder G, Endres K. Transcriptional repression of the ectodomain sheddase ADAM10 by TBX2 and potential implication for Alzheimer's disease. Cell Mol Life Sci 2019; 76:1005-1025. [PMID: 30599067 PMCID: PMC11105458 DOI: 10.1007/s00018-018-2998-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND The ADAM10-mediated cleavage of transmembrane proteins regulates cellular processes such as proliferation or migration. Substrate cleavage by ADAM10 has also been implicated in pathological situations such as cancer or Morbus Alzheimer. Therefore, identifying endogenous molecules, which modulate the amount and consequently the activity of ADAM10, might contribute to a deeper understanding of the enzyme's role in both, physiology and pathology. METHOD To elucidate the underlying cellular mechanism of the TBX2-mediated repression of ADAM10 gene expression, we performed overexpression, RNAi-mediated knockdown and pharmacological inhibition studies in the human neuroblastoma cell line SH-SY5Y. Expression analysis was conducted by e.g. real-time RT-PCR or western blot techniques. To identify the binding region of TBX2 within the ADAM10 promoter, we used luciferase reporter assay on deletion constructs and EMSA/WEMSA experiments. In addition, we analyzed a TBX2 loss-of-function Drosophila model regarding the expression of ADAM10 orthologs by qPCR. Furthermore, we quantified the mRNA level of TBX2 in post-mortem brain tissue of AD patients. RESULTS Here, we report TBX2 as a transcriptional repressor of ADAM10 gene expression: both, the DNA-binding domain and the repression domain of TBX2 were necessary to effect transcriptional repression of ADAM10 in neuronal SH-SY5Y cells. This regulatory mechanism required HDAC1 as a co-factor of TBX2. Transcriptional repression was mediated by two functional TBX2 binding sites within the core promoter sequence (- 315 to - 286 bp). Analysis of a TBX2 loss-of-function Drosophila model revealed that kuzbanian and kuzbanian-like, orthologs of ADAM10, were derepressed compared to wild type. Vice versa, analysis of cortical brain samples of AD-patients, which showed reduced ADAM10 mRNA levels, revealed a 2.5-fold elevation of TBX2, while TBX3 and TBX21 levels were not affected. CONCLUSION Our results characterize TBX2 as a repressor of ADAM10 gene expression and suggest that this regulatory interaction is conserved across tissues and species.
Collapse
Affiliation(s)
- Sven Reinhardt
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Strasse 8, 55131, Mainz, Germany
| | - Florian Schuck
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Strasse 8, 55131, Mainz, Germany
| | - Nicolai Stoye
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Strasse 8, 55131, Mainz, Germany
| | - Tobias Hartmann
- Deutsches Institut für Demenz Prävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Kirrbergerstrasse 1, 66421, Homburg, Saar, Germany
- Experimental Neurology, Saarland University, Kirrbergerstrasse 1, 66421, Homburg, Saar, Germany
| | - Marcus O W Grimm
- Deutsches Institut für Demenz Prävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Kirrbergerstrasse 1, 66421, Homburg, Saar, Germany
- Experimental Neurology, Saarland University, Kirrbergerstrasse 1, 66421, Homburg, Saar, Germany
| | - Gert Pflugfelder
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Becherweg 32, 55128, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Untere Zahlbacher Strasse 8, 55131, Mainz, Germany.
| |
Collapse
|
11
|
Shin H, Reiner DJ. The Signaling Network Controlling C. elegans Vulval Cell Fate Patterning. J Dev Biol 2018; 6:E30. [PMID: 30544993 PMCID: PMC6316802 DOI: 10.3390/jdb6040030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/17/2022] Open
Abstract
EGF, emitted by the Anchor Cell, patterns six equipotent C. elegans vulval precursor cells to assume a precise array of three cell fates with high fidelity. A group of core and modulatory signaling cascades forms a signaling network that demonstrates plasticity during the transition from naïve to terminally differentiated cells. In this review, we summarize the history of classical developmental manipulations and molecular genetics experiments that led to our understanding of the signals governing this process, and discuss principles of signal transduction and developmental biology that have emerged from these studies.
Collapse
Affiliation(s)
- Hanna Shin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
| | - David J Reiner
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA.
- College of Medicine, Texas A & M University, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Wang L, Liu Z, Shi H, Liu J. Two Paralogous Tetraspanins TSP-12 and TSP-14 Function with the ADAM10 Metalloprotease SUP-17 to Promote BMP Signaling in Caenorhabditis elegans. PLoS Genet 2017; 13:e1006568. [PMID: 28068334 PMCID: PMC5261805 DOI: 10.1371/journal.pgen.1006568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/24/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
The highly conserved bone morphogenetic protein (BMP) signaling pathway regulates many developmental and homeostatic processes. While the core components of the BMP pathway have been well studied, much research is needed for understanding the mechanisms involved in the precise spatiotemporal control of BMP signaling in vivo. Here, we provide evidence that two paralogous and evolutionarily conserved tetraspanins, TSP-12 and TSP-14, function redundantly to promote BMP signaling in C. elegans. We further show that the ADAM10 (adisintegrin and metalloprotease 10) ortholog SUP-17 also functions to promote BMP signaling, and that TSP-12 can bind to and promote the cell surface localization of SUP-17. SUP-17/ADAM10 is known to be involved in the ligand-induced proteolytic processing of the Notch receptor. We have evidence that the function of SUP-17, and of TSP-12/TSP-14 in BMP signaling is independent of their roles in Notch signaling. Furthermore, presenilins, core components of the γ-secretase complex involved in processing Notch, do not appear to play a role in BMP signaling. These studies established a new role of the TSP-12/TSP-14/SUP-17 axis in regulating BMP signaling, in addition to their known function in the Notch signaling pathway. We also provide genetic evidence showing that a known BMP signaling modulator, UNC-40/neogenin/DCC, is one of the substrates of SUP-17/ADAM10 in the BMP signaling pathway. Bone morphogenetic protein (BMP) signaling regulates multiple developmental and homeostatic processes. Misregulation of this pathway can cause various diseases, including cancers. Thus, it is essential to understand how BMP signaling is tightly regulated spatiotemporally in vivo. We have identified a highly conserved ADAM (a disintegrin and metalloprotease) protein, SUP-17/ADAM10, as an important factor in modulating BMP signaling in C. elegans. We showed that the proper localization and function of this ADAM protease require two conserved tetraspanin proteins, TSP-12 and TSP-14. We provided genetic evidence showing that one of the substrates of SUP-17/ADAM10 in the BMP signaling pathway is a known BMP signaling modulator, UNC-40/neogenin/DCC. Our studies established a new role of the TSP-12-TSP-14-SUP-17 axis in regulating BMP signaling, in addition to and independent of their known function in the Notch signaling pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Zhiyu Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Herong Shi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
13
|
Toonen JA, Ronchetti A, Sidjanin DJ. A Disintegrin and Metalloproteinase10 (ADAM10) Regulates NOTCH Signaling during Early Retinal Development. PLoS One 2016; 11:e0156184. [PMID: 27224017 PMCID: PMC4880208 DOI: 10.1371/journal.pone.0156184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/10/2016] [Indexed: 01/22/2023] Open
Abstract
ADAM10 and ADAM17 are two closely related members of the ADAM (a disintegrin and metalloprotease) family of membrane-bound sheddases, which proteolytically cleave surface membrane proteins. Both ADAM10 and ADAM17 have been implicated in the proteolytic cleavage of NOTCH receptors and as such regulators of NOTCH signaling. During retinal development, NOTCH signaling facilitates retinal neurogenesis by maintaining progenitor cells in a proliferative state and by mediating retinal cell fates. However, the roles of ADAM10 and ADAM17 in the retina are not well defined. In this study, we set out to clarify the roles of ADAM10 and ADAM17 during early retinal development. The retinal phenotype of conditionally abated Adam17 retinae (Adam17 CKO) did not differ from the controls whereas conditionally ablated Adam10 retinae (Adam10 CKO) exhibited abnormal morphogenesis characterized by the formation of rosettes and a loss of retinal laminae phenotypically similar to morphological abnormalities identified in mice with retinal NOTCH signaling deficiency. Additionally, Adam10 CKO retinae exhibited abnormal neurogenesis characterized by fewer proliferating progenitor cells and greater differentiation of early photoreceptors and retinal ganglion cells. Moreover, constitutive activation of the NOTCH1-intracellular domain (N1-ICD) rescued Adam10 CKO abnormal neurogenesis, as well as abnormal retinal morphology by maintaining retinal cells in the progenitor state. Collectively these findings provide in vivo genetic evidence that ADAM10, and not ADAM17, is indispensable for proper retinal development as a regulator of NOTCH signaling.
Collapse
Affiliation(s)
- Joseph A. Toonen
- Department of Cell Biology, Neurobiology, and Anatomy, 8701 Watertown Plank Rd., Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Adam Ronchetti
- Department of Cell Biology, Neurobiology, and Anatomy, 8701 Watertown Plank Rd., Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - D. J. Sidjanin
- Department of Cell Biology, Neurobiology, and Anatomy, 8701 Watertown Plank Rd., Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Human and Molecular Genetics Center, 8701 Watertown Plank Rd., Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
14
|
Tokumasu Y, Iida A, Wang Z, Ansai S, Kinoshita M, Sehara‐Fujisawa A. ADAM12‐deficient zebrafish exhibit retardation in body growth at the juvenile stage without developmental defects. Dev Growth Differ 2016; 58:409-21. [DOI: 10.1111/dgd.12286] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Yudai Tokumasu
- Department of Growth Regulation Institute for Frontier Medical Sciences Kyoto University Shogo‐in Kawahara‐cho 53 Sakyo‐ku Japan
| | - Atsuo Iida
- Department of Growth Regulation Institute for Frontier Medical Sciences Kyoto University Shogo‐in Kawahara‐cho 53 Sakyo‐ku Japan
| | - Zi Wang
- Department of Growth Regulation Institute for Frontier Medical Sciences Kyoto University Shogo‐in Kawahara‐cho 53 Sakyo‐ku Japan
- Laboratory of Functional Biology Kyoto University Graduate School of Biostudies Japan
| | - Satoshi Ansai
- Division of Applied Biosciences Graduate School of Agriculture Kyoto University Kitashirakawa‐Oiwake‐cho Sakyo‐ku Kyoto 606‐8502 Japan
| | - Masato Kinoshita
- Division of Applied Biosciences Graduate School of Agriculture Kyoto University Kitashirakawa‐Oiwake‐cho Sakyo‐ku Kyoto 606‐8502 Japan
| | - Atsuko Sehara‐Fujisawa
- Department of Growth Regulation Institute for Frontier Medical Sciences Kyoto University Shogo‐in Kawahara‐cho 53 Sakyo‐ku Japan
| |
Collapse
|
15
|
Dong B, Moseley-Alldredge M, Schwieterman AA, Donelson CJ, McMurry JL, Hudson ML, Chen L. EFN-4 functions in LAD-2-mediated axon guidance in Caenorhabditis elegans. Development 2016; 143:1182-91. [PMID: 26903502 DOI: 10.1242/dev.128934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/12/2016] [Indexed: 11/20/2022]
Abstract
During development of the nervous system, growing axons rely on guidance molecules to direct axon pathfinding. A well-characterized family of guidance molecules are the membrane-associated ephrins, which together with their cognate Eph receptors, direct axon navigation in a contact-mediated fashion. InC. elegans, the ephrin-Eph signaling system is conserved and is best characterized for their roles in neuroblast migration during early embryogenesis. This study demonstrates a role for the C. elegans ephrin EFN-4 in axon guidance. We provide both genetic and biochemical evidence that is consistent with the C. elegans divergent L1 cell adhesion molecule LAD-2 acting as a non-canonical ephrin receptor to EFN-4 to promote axon guidance. We also show that EFN-4 probably functions as a diffusible factor because EFN-4 engineered to be soluble can promote LAD-2-mediated axon guidance. This study thus reveals a potential additional mechanism for ephrins in regulating axon guidance and expands the repertoire of receptors by which ephrins can signal.
Collapse
Affiliation(s)
- Bingyun Dong
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Melinda Moseley-Alldredge
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alicia A Schwieterman
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Cory J Donelson
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Jonathan L McMurry
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Martin L Hudson
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Lihsia Chen
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
16
|
Alexander AG, Marfil V, Li C. Use of Caenorhabditis elegans as a model to study Alzheimer's disease and other neurodegenerative diseases. Front Genet 2014; 5:279. [PMID: 25250042 PMCID: PMC4155875 DOI: 10.3389/fgene.2014.00279] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022] Open
Abstract
Advances in research and technology has increased our quality of life, allowed us to combat diseases, and achieve increased longevity. Unfortunately, increased longevity is accompanied by a rise in the incidences of age-related diseases such as Alzheimer’s disease (AD). AD is the sixth leading cause of death, and one of the leading causes of dementia amongst the aged population in the USA. It is a progressive neurodegenerative disorder, characterized by the prevalence of extracellular Aβ plaques and intracellular neurofibrillary tangles, derived from the proteolysis of the amyloid precursor protein (APP) and the hyperphosphorylation of microtubule-associated protein tau, respectively. Despite years of extensive research, the molecular mechanisms that underlie the pathology of AD remain unclear. Model organisms, such as the nematode, Caenorhabditis elegans, present a complementary approach to addressing these questions. C. elegans has many advantages as a model system to study AD and other neurodegenerative diseases. Like their mammalian counterparts, they have complex biochemical pathways, most of which are conserved. Genes in which mutations are correlated with AD have counterparts in C. elegans, including an APP-related gene, apl-1, a tau homolog, ptl-1, and presenilin homologs, such as sel-12 and hop-1. Since the neuronal connectivity in C. elegans has already been established, C. elegans is also advantageous in modeling learning and memory impairments seen during AD. This article addresses the insights C. elegans provide in studying AD and other neurodegenerative diseases. Additionally, we explore the advantages and drawbacks associated with using this model.
Collapse
Affiliation(s)
- Adanna G Alexander
- Department of Biology, City College of New York New York, NY, USA ; Department of Biology, The Graduate Center, City University of New York New York, NY, USA
| | - Vanessa Marfil
- Department of Biology, City College of New York New York, NY, USA
| | - Chris Li
- Department of Biology, City College of New York New York, NY, USA ; Department of Biology, The Graduate Center, City University of New York New York, NY, USA
| |
Collapse
|
17
|
Yan X, Lin J, Talabattula VAN, Mußmann C, Yang F, Wree A, Rolfs A, Luo J. ADAM10 negatively regulates neuronal differentiation during spinal cord development. PLoS One 2014; 9:e84617. [PMID: 24404179 PMCID: PMC3880303 DOI: 10.1371/journal.pone.0084617] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/15/2013] [Indexed: 12/22/2022] Open
Abstract
Members of the ADAM (a disintegrin and metalloprotease) family are involved in embryogenesis and tissue formation via their proteolytic function, cell-cell and cell-matrix interactions. ADAM10 is expressed temporally and spatially in the developing chicken spinal cord, but its function remains elusive. In the present study, we address this question by electroporating ADAM10 specific morpholino antisense oligonucleotides (ADAM10-mo) or dominant-negative ADAM10 (dn-ADAM10) plasmid into the developing chicken spinal cord as well as by in vitro cell culture investigation. Our results show that downregulation of ADAM10 drives precocious differentiation of neural progenitor cells and radial glial cells, resulting in an increase of neurons in the developing spinal cord, even in the prospective ventricular zone. Remarkably, overexpression of the dn-ADAM10 plasmid mutated in the metalloprotease domain (dn-ADAM10-me) mimics the phenotype as found by the ADAM10-mo transfection. Furthermore, in vitro experiments on cultured cells demonstrate that downregulation of ADAM10 decreases the amount of the cleaved intracellular part of Notch1 receptor and its target, and increases the number of βIII-tubulin-positive cells during neural progenitor cell differentiation. Taken together, our data suggest that ADAM10 negatively regulates neuronal differentiation, possibly via its proteolytic effect on the Notch signaling during development of the spinal cord.
Collapse
Affiliation(s)
- Xin Yan
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Rostock, Germany
| | - Juntang Lin
- Key Laboratory for Medical Tissue Regeneration of Henan Province, Xinxiang Medical University, Xinxiang, P.R. China
- Institute of Anatomy I, School of Medicine University of Jena, Jena, Germany
| | | | - Carolin Mußmann
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Rostock, Germany
| | - Fan Yang
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Rostock, Germany
| | - Andreas Wree
- Institute of Anatomy, School of Medicine University of Rostock, Rostock, Germany
| | - Arndt Rolfs
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Rostock, Germany
| | - Jiankai Luo
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Rostock, Germany
- * E-mail:
| |
Collapse
|
18
|
Saraceno C, Musardo S, Marcello E, Pelucchi S, Di Luca M. Modeling Alzheimer's disease: from past to future. Front Pharmacol 2013; 4:77. [PMID: 23801962 PMCID: PMC3685797 DOI: 10.3389/fphar.2013.00077] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/30/2013] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is emerging as the most prevalent and socially disruptive illness of aging populations, as more people live long enough to become affected. Although AD is placing a considerable and increasing burden on society, it represents the largest unmet medical need in neurology, because current drugs improve symptoms, but do not have profound disease-modifying effects. Although AD pathogenesis is multifaceted and difficult to pinpoint, genetic and cell biological studies led to the amyloid hypothesis, which posits that amyloid β (Aβ) plays a pivotal role in AD pathogenesis. Amyloid precursor protein (APP), as well as β- and γ-secretases are the principal players involved in Aβ production, while α-secretase cleavage on APP prevents Aβ deposition. The association of early onset familial AD with mutations in the APP and γ-secretase components provided a potential tool of generating animal models of the disease. However, a model that recapitulates all the aspects of AD has not yet been produced. Here, we face the problem of modeling AD pathology describing several models, which have played a major role in defining critical disease-related mechanisms and in exploring novel potential therapeutic approaches. In particular, we will provide an extensive overview on the distinct features and pros and contras of different AD models, ranging from invertebrate to rodent models and finally dealing with computational models and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Claudia Saraceno
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano Milano, Italy ; Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano Milano, Italy
| | | | | | | | | |
Collapse
|
19
|
Weinstein N, Mendoza L. A network model for the specification of vulval precursor cells and cell fusion control in Caenorhabditis elegans. Front Genet 2013; 4:112. [PMID: 23785384 PMCID: PMC3682179 DOI: 10.3389/fgene.2013.00112] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/28/2013] [Indexed: 01/21/2023] Open
Abstract
The vulva of Caenorhabditis elegans has been long used as an experimental model of cell differentiation and organogenesis. While it is known that the signaling cascades of Wnt, Ras/MAPK, and NOTCH interact to form a molecular network, there is no consensus regarding its precise topology and dynamical properties. We inferred the molecular network, and developed a multivalued synchronous discrete dynamic model to study its behavior. The model reproduces the patterns of activation reported for the following types of cell: vulval precursor, first fate, second fate, second fate with reversed polarity, third fate, and fusion fate. We simulated the fusion of cells, the determination of the first, second, and third fates, as well as the transition from the second to the first fate. We also used the model to simulate all possible single loss- and gain-of-function mutants, as well as some relevant double and triple mutants. Importantly, we associated most of these simulated mutants to multivulva, vulvaless, egg-laying defective, or defective polarity phenotypes. The model shows that it is necessary for RAL-1 to activate NOTCH signaling, since the repression of LIN-45 by RAL-1 would not suffice for a proper second fate determination in an environment lacking DSL ligands. We also found that the model requires the complex formed by LAG-1, LIN-12, and SEL-8 to inhibit the transcription of eff-1 in second fate cells. Our model is the largest reconstruction to date of the molecular network controlling the specification of vulval precursor cells and cell fusion control in C. elegans. According to our model, the process of fate determination in the vulval precursor cells is reversible, at least until either the cells fuse with the ventral hypoderm or divide, and therefore the cell fates must be maintained by the presence of extracellular signals.
Collapse
Affiliation(s)
| | - Luis Mendoza
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de MéxicoMexico City, México
| |
Collapse
|
20
|
Hartenstein V, Wodarz A. Initial neurogenesis in Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 2:701-21. [PMID: 24014455 DOI: 10.1002/wdev.111] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Early neurogenesis comprises the phase of nervous system development during which neural progenitor cells are born. In early development, the embryonic ectoderm is subdivided by a conserved signaling mechanism into two main domains, the epidermal ectoderm and the neurectoderm. Subsequently, cells of the neurectoderm are internalized and form a cell layer of proliferating neural progenitors. In vertebrates, the entire neurectoderm folds into the embryo to give rise to the neural tube. In Drosophila and many other invertebrates, a subset of neurectodermal cells, called neuroblasts (NBs), delaminates and forms the neural primordium inside the embryo where they divide in an asymmetric, stem cell-like mode. The remainder of the neurectodermal cells that stay behind at the surface loose their neurogenic potential and later give rise to the ventral part of the epidermis. The genetic and molecular analysis of the mechanisms controlling specification and proliferation of NBs in the Drosophila embryo, which played a significant part in pioneering the field of modern developmental neurobiology, represents the topic of this review.
Collapse
Affiliation(s)
- Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
21
|
Xu G, Wei S, White JM, DeSimone DW. Identification and characterization of ADAM41, a novel ADAM metalloproteinase in Xenopus. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2012; 56:333-9. [PMID: 22811267 DOI: 10.1387/ijdb.113444gx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The ADAM family of transmembrane metalloproteinases has important functions in fertilization, development and disease, and is widely distributed throughout the Metazoa. In this study, we identified a novel ADAM protein in Xenopus tropicalis (X. tropicalis) with closest overall sequence similarity to the Xenopus ADAM10 protein. Based on comparisons of available sequence information, putative orthologs of this ADAM (which we designate ADAM41) are identified in several other vertebrate species including non-placental mammals, but absent from placental mammals and aves. ADAM41 mRNA is maternally deposited in X. tropicalis with subsequent zygotic expression detected in the neural plate at neurula stages. Antisense morpholino knockdown of ADAM41 results in a delay in early neuronal marker expression, which can be rescued by a non-targeted ADAM41 transcript. Thus, ADAM41 is likely required for maintaining proper timing of neurogenesis in X. tropicalis.
Collapse
Affiliation(s)
- Guofeng Xu
- Department of Cell Biology and the Morphogenesis and Regenerative Medicine Institute, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Proteolytic enzymes belonging to the A Disintegin And Metalloproteinase (ADAM) family are able to cleave transmembrane proteins close to the cell surface, in a process referred to as ectodomain shedding. Substrates for ADAMs include growth factors, cytokines, chemokines and adhesion molecules, and, as such, many ADAM proteins play crucial roles in cell-cell adhesion, extracellular and intracellular signaling, cell differentiation and cell proliferation. In this Review, we summarize the fascinating roles of ADAMs in embryonic and adult tissue development in both vertebrates and invertebrates.
Collapse
Affiliation(s)
- Silvio Weber
- Heart Research Centre Göttingen, Universitaetsmedizin Göttingen, Department of Cardiology and Pneumology, Georg-August-University Göttingen, Germany
| | | |
Collapse
|
23
|
Abstract
Many neurons have limited capacity to regenerate their axons after injury. Neurons in the mammalian central nervous system do not regenerate, and even neurons in the peripheral nervous system often fail to regenerate to their former targets. This failure is likely due in part to pathways that actively restrict regeneration; however, only a few factors that limit regeneration are known. Here, using single-neuron analysis of regeneration in vivo, we show that Notch/lin-12 signaling inhibits the regeneration of mature C. elegans neurons. Notch signaling suppresses regeneration by acting autonomously in the injured cell to prevent growth cone formation. The metalloprotease and gamma-secretase cleavage events that lead to Notch activation during development are also required for its activity in regeneration. Furthermore, blocking Notch activation immediately after injury improves regeneration. Our results define a postdevelopmental role for the Notch pathway as a repressor of axon regeneration in vivo.
Collapse
Affiliation(s)
- Rachid El Bejjani
- Department of Genetics, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 295 Congress Avenue, New Haven, CT 06510, USA
| | | |
Collapse
|
24
|
Abstract
Regulated intramembrane proteolysis (RIP) is a highly conserved signaling paradigm whereby membrane-bound signaling proteins are cleaved in their transmembrane region and then released into the cytoplasm to act as signaling molecules. In most if not all cases intramembrane cleavage is preceded and regulated by a membrane proximal cleavage step called 'ectodomain shedding'. Here we will review the role of ectodomain shedding in RIP of the NOTCH signaling pathway, a highly conserved cell-cell communication pathway that mediates cell fate decisions during development and in adult tissues.
Collapse
|
25
|
Abstract
Notch signaling is integral to a large number of developmental and homeostasis events, and either gain or loss of Notch signaling results in a wide range of defects. Notch must be processed by several proteases, including a member of the ADAM (a disintegrin and metalloprotease) family to mediate downstream signaling. Until recently, interactions of Notch with specific ADAMs in different contexts were unclear. ADAM10 is now known to be specifically essential for development and homeostasis of mouse epidermis and cardiovascular structures, and ADAM17 may not be able to fully replace ADAM10 in these contexts. However, Notch from T-cell acute lymphoblastic leukemia (T-ALL) patients can be cleaved by both ADAMs 10 and 17. Studies have revealed that ADAM10 is necessary for Notch processing when Notch is activated by a ligand, while ADAM17 is the major protease for processing Notch that is activated independently of ligand in both flies and mammals.
Collapse
Affiliation(s)
- Laura M Christian
- Department of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
26
|
Abstract
The Notch pathway is a highly conserved signaling pathway in multicellular eukaryotes essential in controlling spatial patterning, morphogenesis and homeostasis in embryonic and adult tissues. Notch proteins coordinate cell-cell communication through receptor-ligand interactions between adjacent cells. Notch signaling is frequently deregulated by oncogenic mutation or overexpression in many cancer types. Notch activity is controlled by three sequential cleavage steps leading to ectodomain shedding and transcriptional activation. Here we review the key regulatory steps in the activation of Notch, from receptor maturation to receptor activation (HIT) via a rate-limiting proteolytic cascade (RUN) in the context of species-specific differences.
Collapse
Affiliation(s)
- G. van Tetering
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - M. Vooijs
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Radiotherapy (MAASTRO)/GROW School for Oncology and Developmental Biology, University of Maastricht, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
27
|
Genetics of extracellular matrix remodeling during organ growth using the Caenorhabditis elegans pharynx model. Genetics 2010; 186:969-82. [PMID: 20805556 DOI: 10.1534/genetics.110.120519] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The organs of animal embryos are typically covered with an extracellular matrix (ECM) that must be carefully remodeled as these organs enlarge during post-embryonic growth; otherwise, their shape and functions may be compromised. We previously described the twisting of the Caenorhabditis elegans pharynx (here called the Twp phenotype) as a quantitative mutant phenotype that worsens as that organ enlarges during growth. Mutations previously known to cause pharyngeal twist affect membrane proteins with large extracellular domains (DIG-1 and SAX-7), as well as a C. elegans septin (UNC-61). Here we show that two novel alleles of the C. elegans papilin gene, mig-6(et4) and mig-6(sa580), can also cause the Twp phenotype. We also show that overexpression of the ADAMTS protease gene mig-17 can suppress the pharyngeal twist in mig-6 mutants and identify several alleles of other ECM-related genes that can cause or influence the Twp phenotype, including alleles of fibulin (fbl-1), perlecan (unc-52), collagens (cle-1, dpy-7), laminins (lam-1, lam-3), one ADAM protease (sup-17), and one ADAMTS protease (adt-1). The Twp phenotype in C. elegans is easily monitored using light microscopy, is quantitative via measurements of the torsion angle, and reveals that ECM components, metalloproteinases, and ECM attachment molecules are important for this organ to retain its correct shape during post-embryonic growth. The Twp phenotype is therefore a promising experimental system to study ECM remodeling and diseases.
Collapse
|
28
|
Wei S, Whittaker CA, Xu G, Bridges LC, Shah A, White JM, Desimone DW. Conservation and divergence of ADAM family proteins in the Xenopus genome. BMC Evol Biol 2010; 10:211. [PMID: 20630080 PMCID: PMC3055250 DOI: 10.1186/1471-2148-10-211] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 07/14/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Members of the disintegrin metalloproteinase (ADAM) family play important roles in cellular and developmental processes through their functions as proteases and/or binding partners for other proteins. The amphibian Xenopus has long been used as a model for early vertebrate development, but genome-wide analyses for large gene families were not possible until the recent completion of the X. tropicalis genome sequence and the availability of large scale expression sequence tag (EST) databases. In this study we carried out a systematic analysis of the X. tropicalis genome and uncovered several interesting features of ADAM genes in this species. RESULTS Based on the X. tropicalis genome sequence and EST databases, we identified Xenopus orthologues of mammalian ADAMs and obtained full-length cDNA clones for these genes. The deduced protein sequences, synteny and exon-intron boundaries are conserved between most human and X. tropicalis orthologues. The alternative splicing patterns of certain Xenopus ADAM genes, such as adams 22 and 28, are similar to those of their mammalian orthologues. However, we were unable to identify an orthologue for ADAM7 or 8. The Xenopus orthologue of ADAM15, an active metalloproteinase in mammals, does not contain the conserved zinc-binding motif and is hence considered proteolytically inactive. We also found evidence for gain of ADAM genes in Xenopus as compared to other species. There is a homologue of ADAM10 in Xenopus that is missing in most mammals. Furthermore, a single scaffold of X. tropicalis genome contains four genes encoding ADAM28 homologues, suggesting genome duplication in this region. CONCLUSIONS Our genome-wide analysis of ADAM genes in X. tropicalis revealed both conservation and evolutionary divergence of these genes in this amphibian species. On the one hand, all ADAMs implicated in normal development and health in other species are conserved in X. tropicalis. On the other hand, some ADAM genes and ADAM protease activities are absent, while other novel ADAM proteins in this species are predicted by this study. The conservation and unique divergence of ADAM genes in Xenopus probably reflect the particular selective pressures these amphibian species faced during evolution.
Collapse
Affiliation(s)
- Shuo Wei
- Department of Cell Biology and the Morphogenesis and Regenerative Medicine Institute, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Dunn CD, Sulis ML, Ferrando AA, Greenwald I. A conserved tetraspanin subfamily promotes Notch signaling in Caenorhabditis elegans and in human cells. Proc Natl Acad Sci U S A 2010; 107:5907-12. [PMID: 20220101 PMCID: PMC2851858 DOI: 10.1073/pnas.1001647107] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cytosolic domain of Notch is a membrane-tethered transcription factor. Ligand binding ultimately leads to gamma-secretase cleavage within the transmembrane domain, allowing the intracellular domain to translocate to the nucleus and activate target gene transcription. Constitutive Notch signaling has been associated with human cancers such as T cell acute lymphoblastic leukemia (T-ALL). As tetraspanins have been implicated in many different signaling processes, we assessed their potential contribution to Notch signaling. We used a genetic assay in Caenorhabditis elegans to identify TSP-12 as a positive factor for Notch activity in several cellular contexts. Then, using a cell culture system, we showed that two human TSP-12 orthologs, TSPAN33 and TSPAN5, promote Notch activity and are likely to act at the gamma-secretase cleavage step. We also acquired evidence for functional redundancy among tetraspanins in both C. elegans and human cells. Selective inhibition of tetraspanins may constitute an anti-NOTCH therapeutic approach to reduce gamma-secretase activity.
Collapse
Affiliation(s)
- Cory D. Dunn
- Howard Hughes Medical Institute
- Department of Biochemistry and Molecular Biophysics
| | - Maria Luisa Sulis
- Division of Pediatric Oncology, Department of Pediatrics
- Institute for Cancer Genetics
| | - Adolfo A. Ferrando
- Division of Pediatric Oncology, Department of Pediatrics
- Institute for Cancer Genetics
- Department of Pathology, and
| | - Iva Greenwald
- Howard Hughes Medical Institute
- Department of Biochemistry and Molecular Biophysics
- Department of Genetics and Development, Columbia University College of Physicians and Surgeons New York, NY 10032
| |
Collapse
|
30
|
Ewald CY, Li C. Understanding the molecular basis of Alzheimer's disease using a Caenorhabditis elegans model system. Brain Struct Funct 2010; 214:263-83. [PMID: 20012092 PMCID: PMC3902020 DOI: 10.1007/s00429-009-0235-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 11/17/2009] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the major cause of dementia in the United States. At the cellular level, the brains of AD patients are characterized by extracellular dense plaques and intracellular neurofibrillary tangles whose major components are the beta-amyloid peptide and tau, respectively. The beta-amyloid peptide is a cleavage product of the amyloid precursor protein (APP); mutations in APP have been correlated with a small number of cases of familial Alzheimer's disease. APP is the canonical member of the APP family, whose functions remain unclear. The nematode Caenorhabditis elegans, one of the premier genetic workhorses, is being used in a variety of ways to address the functions of APP and determine how the beta-amyloid peptide and tau can induce toxicity. First, the function of the C. elegans APP-related gene, apl-1, is being examined. Although different organisms may use APP and related proteins, such as APL-1, in different functional contexts, the pathways in which they function and the molecules with which they interact are usually conserved. Second, components of the gamma-secretase complex and their respective functions are being revealed through genetic analyses in C. elegans. Third, to address questions of toxicity, onset of degeneration, and protective mechanisms, different human beta-amyloid peptide and tau variants are being introduced into C. elegans and the resultant transgenic lines examined. Here, we summarize how a simple system such as C. elegans can be used as a model to understand APP function and suppression of beta-amyloid peptide and tau toxicity in higher organisms.
Collapse
Affiliation(s)
- Collin Y. Ewald
- Graduate Center and Department of Biology, City College of the City University of New York, MR526, 160 Convent Avenue, New York, NY 10031, USA
| | - Chris Li
- Graduate Center and Department of Biology, City College of the City University of New York, MR526, 160 Convent Avenue, New York, NY 10031, USA
| |
Collapse
|
31
|
Trang SH, Joyner DE, Damron TA, Aboulafia AJ, Randall RL. Potential for functional redundancy in EGF and TGFalpha signaling in desmoid cells: a cDNA microarray analysis. Growth Factors 2010; 28:10-23. [PMID: 20092031 DOI: 10.3109/08977190903299387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genes that replace or duplicate the function of other genes are considered functionally redundant. In this cDNA microarray study, using an Agilent microarray platform and GeneSifter analysis software, we evaluated (1) the degree of downstream transcriptional redundancy and (2) the level of genetic uniqueness apparent in desmoid tumor cells stimulated in vitro for 3 h or for 24 h with 100 ng/ml of exogenous recombinant human EGF (rhEGF) or with recombinant human transforming growth factor alpha (rhTGFalpha). Our intent was to identify genes costimulated, or genes unique to, desmoid cells stimulated in vitro with rhEGF and rhTGFalpha. This experimental approach demonstrated a 55% transcriptional redundancy in the number of desmoid genes significantly upregulated or downregulated following 3 h of stimulation with rhEGF or with rhTGFalpha, and a 65% transcriptional redundancy following 24 h of growth factor stimulation. Approximately 150 genes costimulated by rhEGF and rhTGFalpha were identified. This study suggests that EGF and TGFalpha retain some level of functional redundancy, possibly resulting from their divergence from a common ancestral gene.
Collapse
Affiliation(s)
- Sylvia H Trang
- SARC Laboratory, Sarcoma Services, Department of Orthopaedics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
32
|
van Tetering G, van Diest P, Verlaan I, van der Wall E, Kopan R, Vooijs M. Metalloprotease ADAM10 is required for Notch1 site 2 cleavage. J Biol Chem 2009; 284:31018-27. [PMID: 19726682 DOI: 10.1074/jbc.m109.006775] [Citation(s) in RCA: 217] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Notch signaling is controlled by ligand binding, which unfolds a negative control region to induce proteolytic cleavage of the receptor. First, a membrane-proximal cleavage is executed by a metalloprotease, removing the extracellular domain. This allows gamma-secretase to execute a second cleavage within the Notch transmembrane domain, which releases the intracellular domain to enter the nucleus. Here we show that the ADAM10 metalloprotease Kuzbanian, but not ADAM17/tumor necrosis factor alpha-converting enzyme, plays an essential role in executing ligand-induced extracellular cleavage at site 2 (S2) in cells and localizes this step to the plasma membrane. Importantly, genetic or pharmacological inhibition of metalloproteases still allowed extracellular cleavage of Notch, indicating the presence of unknown proteases with the ability to cleave at S2. Gain of function mutations identified in human cancers and in model organisms that map to the negative control region alleviate the requirement for ligand binding for extracellular cleavage to occur. Because cancer-causing Notch1 mutations also depend on (rate-limiting) S2 proteolysis, the identity of these alternative proteases has important implications for understanding Notch activation in normal and cancer cells.
Collapse
Affiliation(s)
- Geert van Tetering
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Notch signaling requires a series of proteolytic cleavage events to release the Notch intracellular domain (NICD) that functions directly in signal transduction. The Notch receptor is locked down in a protease-resistant state by a negative regulatory region (NRR) that protects an ADAM (a disintegrin and metalloprotease) cleavage site. Engagement with ligand-bearing cells induces global conformational movements in Notch that unfold the NRR structure to expose the ADAM cleavage site and initiate proteolytic activation. Although both ADAM10 and ADAM17 have been reported to cleave Notch to facilitate NICD release by gamma-secretase, the relevant ADAM has remained controversial. Our study provides new insight into this conflict, as we find that although Notch1 (N1) is a substrate for both ADAM10 and ADAM17, the particular ADAM required for receptor activation is context dependent. Specifically, ADAM10 was absolutely required for N1 signaling induced by ligands, while signaling independent of ligands required ADAM17. In contrast to the strict and differential use of ADAM10 and ADAM17 in normal and dysregulated signaling, respectively, both proteases participated in signaling intrinsic to N1 mutations associated with leukemia. We propose that in addition to exposing the ADAM cleavage site, activating N1 conformational changes facilitate selective cleavage by specific proteases.
Collapse
|
34
|
Alfandari D, McCusker C, Cousin H. ADAM function in embryogenesis. Semin Cell Dev Biol 2009; 20:153-63. [PMID: 18935966 PMCID: PMC2693894 DOI: 10.1016/j.semcdb.2008.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/22/2008] [Accepted: 09/24/2008] [Indexed: 12/22/2022]
Abstract
Cleavage of proteins inserted into the plasma membrane (shedding) is an essential process controlling many biological functions including cell signaling, cell adhesion and migration as well as proliferation and differentiation. ADAM surface metalloproteases have been shown to play an essential role in these processes. Gene inactivation during embryonic development have provided evidence of the central role of ADAM proteins in nematodes, flies, frogs, birds and mammals. The relative contribution of four subfamilies of ADAM proteins to developmental processes is the focus of this review.
Collapse
Affiliation(s)
- Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Paige lab. Rm. 203, 161 Holdsworth Way, Amherst, MA 01003, United States.
| | | | | |
Collapse
|
35
|
Transcriptome analysis of the zebrafish mind bomb mutant. Mol Genet Genomics 2008; 281:77-85. [PMID: 19005681 DOI: 10.1007/s00438-008-0395-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 10/16/2008] [Indexed: 12/31/2022]
Abstract
Mind bomb (Mib) facilitates Notch signaling pathway by promoting the endocytosis of Notch ligand. The zebrafish mib ( ta52b ) mutant has a defect in its ubiquitin ligase activity which is necessary to inhibit the neurogenesis, resulting in a neuronal hyperplasia. Several genes regulated in the mib ( ta52b ) mutant have been well established, however, there were relatively few reports about the transcriptome profile. To identify the genes differentially expressed in the mib ( ta52b ) mutant, genome-wide analysis was performed using serial analysis of gene expression. Three hundred and thirty-five transcripts were identified whose expressions were significantly altered in the mib ( ta52b ) mutant as compared with the wild-type. Interestingly, it was suggested that the mib ( ta52b ) mutation may affect not only neurogenesis but also mesoderm development. These results provide new insights into Notch signaling pathway.
Collapse
|
36
|
Fiky AE, Pioli P, Azam A, Yoo K, Nastiuk KL, Krolewski JJ. Nuclear transit of the intracellular domain of the interferon receptor subunit IFNaR2 requires Stat2 and Irf9. Cell Signal 2008; 20:1400-8. [PMID: 18456457 PMCID: PMC2494602 DOI: 10.1016/j.cellsig.2008.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 03/17/2008] [Indexed: 01/15/2023]
Abstract
Regulated intramembrane proteolysis (RIP) is the primary signaling mechanism for some receptors, such as Notch and the amyloid precursor protein. In addition, some receptor type tyrosine kinases, such as HER4, are able to signal via both kinase activation and regulated receptor proteolysis. Previously, we showed that the IFNaR2 subunit of the type I interferon receptor can be cleaved in a two step process that resembles RIP and that the IFNaR2 intracellular domain (IFNaR2-ICD) can mediate gene transcription in a Stat2 dependent manner. Here, we demonstrate that IFNaR2-ICD, Stat2 and Irf9 form a ternary complex. Furthermore, Stat2 and Irf9 are required for the nuclear transit of a GFP-linked IFNaR2-ICD construct (GFP-ICD). Additional experiments monitoring the nuclear localization of GFP-ICD demonstrate that Stat2 serves an adaptor role, mediating the interaction between the IFNaR2-ICD and Irf9, while the bipartite nuclear localization signal within Irf9 is the primary determinant driving nuclear transit of the ICD containing complex. Overall, the data suggest that liberation of the IFNaR2-ICD by regulated proteolysis could trigger a novel mechanism for moving the transcription factor Stat2 to the nucleus.
Collapse
Affiliation(s)
- Ashraf El Fiky
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| | - Pete Pioli
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| | - Arif Azam
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| | - Kiwon Yoo
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| | - Kent L. Nastiuk
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| | - John J. Krolewski
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, IRVINE, Irvine, CA 92679
- Chao Family Comprehensive Cancer Center, School of Medicine, University of California, IRVINE, Irvine, CA 92679
| |
Collapse
|
37
|
Caffaro CE, Hirschberg CB, Berninsone PM. Functional redundancy between two Caenorhabditis elegans nucleotide sugar transporters with a novel transport mechanism. J Biol Chem 2007; 282:27970-5. [PMID: 17652078 DOI: 10.1074/jbc.m704485200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transporters of nucleotide sugars regulate the availability of these substrates required for glycosylation reactions in the lumen of the Golgi apparatus and play an important role in the development of multicellular organisms. Caenorhabditis elegans has seven different sugars in its glycoconjugates, although 18 putative nucleotide sugar transporters are encoded in the genome. Among these, SQV-7, SRF-3, and CO3H5.2 exhibit partially overlapping substrate specificity and expression patterns. We now report evidence of functional redundancy between transporters CO3H5.2 and SRF-3. Reducing the activity of the CO3H5.2 gene product by RNA interference (RNAi) in SRF-3 mutants results in oocyte accumulation and abnormal gonad morphology, whereas comparable RNAi treatment of wild type or RNAi hypersensitive C. elegans strains does not cause detectable defects. We hypothesize this genetic enhancement to be a mechanism to ensure adequate glycoconjugate biosynthesis required for normal tissue development in multicellular organisms. Furthermore, we show that transporters SRF-3 and CO3H5.2, which are closely related in the phylogenetic tree, share a simultaneous and independent substrate transport mechanism that is different from the competitive one previously demonstrated for transporter SQV-7, which shares a lower amino acid sequence identity with CO3H5.2 and SRF-3. Therefore, different mechanisms for transporting multiple nucleotide sugars may have evolved parallel to transporter amino acid divergence.
Collapse
Affiliation(s)
- Carolina E Caffaro
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
38
|
Li N, Boyd K, Dempsey PJ, Vignali DAA. Non-Cell Autonomous Expression of TNF-α-Converting Enzyme ADAM17 Is Required for Normal Lymphocyte Development. THE JOURNAL OF IMMUNOLOGY 2007; 178:4214-21. [PMID: 17371977 DOI: 10.4049/jimmunol.178.7.4214] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TNF-alpha converting enzyme (TACE; ADAM17), a member of the ADAM (a disintegrin and metalloprotease) family of metalloproteases, has been shown to cleave a wide variety of cell surface proteins of immunological importance. Due to the broad expression of TACE and the early postnatal lethality of TACE-deficient mice, it has been difficult to assess the role of TACE in lymphocyte development. Indeed, it is not known whether hemopoietic and/or nonhemopoietic expression of TACE is required for normal lymphocyte development. In the current study, we analyzed the lymphoid system of tace(DeltaZn/DeltaZn) mice and tace(DeltaZn/DeltaZn) bone marrow RAG1(-/-) recipients. Our results clearly show that nonlymphocyte expression of TACE is required for normal lymphocyte development and lymphoid organ structure. Lack of TACE function resulted in a partial block in T cell development at the double-negative 4:double-positive transition in the thymus, a loss of B cell development/maturation in the spleen, and a lack of B cell follicle and germinal center formation in the spleen. Thus, TACE serves as a lymphocyte extrinsic factor that is essential for normal T development and peripheral B cell maturation.
Collapse
Affiliation(s)
- Nianyu Li
- Department of Immunology, St. Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) is a member of low-density lipoprotein receptor (LDLR) family which cooperates with Frizzled receptors to transduce the canonical Wnt signal. As a critical component of the canonical Wnt pathway, LRP6 is essential for appropriate brain development, however, the mechanism by which LRP6 facilitates Wnt canonical signaling has not been fully elucidated. Interestingly, LRP6 which lacks its extracellular domain can constitutively activate TCF/LEF and potentiate the Wnt signal. Further, the free cytosolic tail of LRP6 interacts directly with glycogen synthase kinase (GSK3) and inhibits GSK3's activity in the Wnt canonical pathway which results in increased TCF/LEF activation. However, whether these truncated forms of LRP6 are physiologically relevant is unclear. Recent studies have shown that other members of the LDLR family undergo gamma-secretase dependent regulated intramembrane proteolysis (RIP). Using independent experimental approaches, we show that LRP6 also undergoes RIP. The extracellular domain of LRP6 is shed and released into the surrounding milieu and the cytoplasmic tail is cleaved by gamma-secretase-like activity to release the intracellular domain. Furthermore, protein kinase C, Wnt 3a and Dickkopf-1 modulate this process. These findings suggest a novel mechanism for LRP6 in Wnt signaling: induction of ectodomain shedding of LRP6, followed by the gamma-secretase involved proteolytic releasing its intracellular domain (ICD) which then binds to GSK3 inhibiting its activity and thus activates the canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Kaihong Mi
- Department of Psychiatry, University of Alabama at Birmingham, Birmingham, Alabama 35294-0017, USA
| | | |
Collapse
|
40
|
Doroquez DB, Rebay I. Signal integration during development: mechanisms of EGFR and Notch pathway function and cross-talk. Crit Rev Biochem Mol Biol 2007; 41:339-85. [PMID: 17092823 DOI: 10.1080/10409230600914344] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Metazoan development relies on a highly regulated network of interactions between conserved signal transduction pathways to coordinate all aspects of cell fate specification, differentiation, and growth. In this review, we discuss the intricate interplay between the epidermal growth factor receptor (EGFR; Drosophila EGFR/DER) and the Notch signaling pathways as a paradigm for signal integration during development. First, we describe the current state of understanding of the molecular architecture of the EGFR and Notch signaling pathways that has resulted from synergistic studies in vertebrate, invertebrate, and cultured cell model systems. Then, focusing specifically on the Drosophila eye, we discuss how cooperative, sequential, and antagonistic relationships between these pathways mediate the spatially and temporally regulated processes that generate this sensory organ. The common themes underlying the coordination of the EGFR and Notch pathways appear to be broadly conserved and should, therefore, be directly applicable to elucidating mechanisms of information integration and signaling specificity in vertebrate systems.
Collapse
Affiliation(s)
- David B Doroquez
- Department of Biology, Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
41
|
Abstract
A small number of signalling pathways are used iteratively to regulate cell fates, cell proliferation and cell death in development. Notch is the receptor in one such pathway, and is unusual in that most of its ligands are also transmembrane proteins; therefore signalling is restricted to neighbouring cells. Although the intracellular transduction of the Notch signal is remarkably simple, with no secondary messengers, this pathway functions in an enormous diversity of developmental processes and its dysfunction is implicated in many cancers.
Collapse
Affiliation(s)
- Sarah J Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| |
Collapse
|
42
|
Tousseyn T, Jorissen E, Reiss K, Hartmann D. (Make) stick and cut loose--disintegrin metalloproteases in development and disease. ACTA ACUST UNITED AC 2006; 78:24-46. [PMID: 16622847 DOI: 10.1002/bdrc.20066] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
"A disintegrin and metalloprotease" (ADAM) proteases form a still growing family of about 40 type 1 transmembrane proteins. They are defined by a common modular ectodomain architecture that combines cell deadhesion/adhesion and fusion motifs (disintegrin and cysteine-rich domains), with a Zn-protease domain capped by a large prodomain. Their ectodomain thus strikingly resembles snake venom disintegrin proteases, which by combined integrin blocking and extracellular proteolysis, can cause extensive tissue damage after snake bites. A surprisingly large proportion (13 ADAMs) is exclusively expressed in the male gonads, and only a minority can be found throughout all tissues. As predicted by their amino acid sequence, a major proportion of this family has not maintained a functional protease domain, most probably rendering them into pure adhesion and/or fusion proteins. For most ADAMs, the respective key function has remained elusive. Despite their overall conserved ectodomain structure, ADAMs appear to be subdivided into those with a predominant role in direct adhesion (e.g., ADAMs 1, 2, and 3) and those mainly acting as proteases (e.g., ADAMs 10 and 17). Only for a few of them are functions of more than one domain documented (e.g., ADAM9 in cell fusion and proteolysis). Several ADAMs exist in both membrane-resident and secreted isoforms; the functional significance of this dichotomy is in most cases still unclear. Knockout phenotypes have been informative only in a few cases (ADAMs 1, 2, 10, 12, 15, 17, and 19) and are mainly related to their protease function. A common denominator of ADAM-mediated proteolysis is the ectodomain shedding of a broad spectrum of substrates, including paracrine growth factors like epidermal growth factor receptor (EGFR) ligands, cell adhesion molecules like CD44 or cadherins, and the initiation of regulated intramembrane proteolysis (RIP), whereby the transmembrane fragment of the respective substrate is further cleaved by an intramembrane cleaving protease to release an intracellular domain acting as a nuclear transcription regulator. Most ADAMs feature a significant overlap of substrate specificities, explaining why an inactivation of individual ADAMs only rarely causes major phenotypes.
Collapse
Affiliation(s)
- Thomas Tousseyn
- Laboratory for Neuronal Cell Biology and Gene Transfer, Department for Human Genetics, K.U. Leuven and Flanders Interuniversity Institute for Biotechnology, Leuven/Flanders, Belgium
| | | | | | | |
Collapse
|