1
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2024; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
2
|
Qian X, Chen K, Chen L, Song H, Zhang Z. Presenilin is involved in larval-pupal metamorphosis development of Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 109:e21855. [PMID: 34811799 DOI: 10.1002/arch.21855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Disruption of the presenilin (ps) genes are the major genetic cause of familial Alzheimer's disease. The silkworm, Bombyx mori (B. mori), is an important model insect. The ps homologue gene in B. mori was identified and characterized. However, the role of ps in B. mori was poorly understood. Here, we found that Bmps was ubiquitously expressed in all the tested tissues during metamorphosis. In the current study, loss-of-function analysis of Bmps was performed by the binary transgenic CRISPR/cas9 system. Compared with the wild type, the developmental time of ∆Bmps animals were significantly delayed. In addition, ∆Bmps showed abnormal appendage including antenna, leg, wing and eye during pupal and adult stages. RNA-seq analysis indicated that apoptosis and proliferation related pathways were affected in ∆Bmps. Moreover, the Hippo pathway was affected by Bmps depletion in brain and wing disc. Our results suggest that PS is essential for maintaining the dynamic balance of apoptosis and proliferation during metamorphosis.
Collapse
Affiliation(s)
- Xiaoran Qian
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Kai Chen
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
| | - Lijuan Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Hongsheng Song
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhongjie Zhang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
3
|
Jia L, Piña-Crespo J, Li Y. Restoring Wnt/β-catenin signaling is a promising therapeutic strategy for Alzheimer's disease. Mol Brain 2019; 12:104. [PMID: 31801553 PMCID: PMC6894260 DOI: 10.1186/s13041-019-0525-5] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/26/2019] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is an aging-related neurological disorder characterized by synaptic loss and dementia. Wnt/β-catenin signaling is an essential signal transduction pathway that regulates numerous cellular processes including cell survival. In brain, Wnt/β-catenin signaling is not only crucial for neuronal survival and neurogenesis, but it plays important roles in regulating synaptic plasticity and blood-brain barrier integrity and function. Moreover, activation of Wnt/β-catenin signaling inhibits amyloid-β production and tau protein hyperphosphorylation in the brain. Critically, Wnt/β-catenin signaling is greatly suppressed in AD brain via multiple pathogenic mechanisms. As such, restoring Wnt/β-catenin signaling represents a unique opportunity for the rational design of novel AD therapies.
Collapse
Affiliation(s)
- Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.,Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, 361102, China
| | - Juan Piña-Crespo
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
4
|
Suresh J, Harmston N, Lim KK, Kaur P, Jin HJ, Lusk JB, Petretto E, Tolwinski NS. An embryonic system to assess direct and indirect Wnt transcriptional targets. Sci Rep 2017; 7:11092. [PMID: 28894169 PMCID: PMC5593962 DOI: 10.1038/s41598-017-11519-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023] Open
Abstract
During animal development, complex signals determine and organize a vast number of tissues using a very small number of signal transduction pathways. These developmental signaling pathways determine cell fates through a coordinated transcriptional response that remains poorly understood. The Wnt pathway is involved in a variety of these cellular functions, and its signals are transmitted in part through a β-catenin/TCF transcriptional complex. Here we report an in vivo Drosophila assay that can be used to distinguish between activation, de-repression and repression of transcriptional responses, separating upstream and downstream pathway activation and canonical/non-canonical Wnt signals in embryos. We find specific sets of genes downstream of both β-catenin and TCF with an additional group of genes regulated by Wnt, while the non-canonical Wnt4 regulates a separate cohort of genes. We correlate transcriptional changes with phenotypic outcomes of cell differentiation and embryo size, showing our model can be used to characterize developmental signaling compartmentalization in vivo.
Collapse
Affiliation(s)
- Jahnavi Suresh
- Yale-NUS College, 12 College Ave West, #01- 201, Singapore, 138610, Republic of Singapore
| | - Nathan Harmston
- Duke-NUS Medical School, 8 College Road, 169857, Singapore, Republic of Singapore
| | - Ka Keat Lim
- Duke-NUS Medical School, 8 College Road, 169857, Singapore, Republic of Singapore
| | - Prameet Kaur
- Yale-NUS College, 12 College Ave West, #01- 201, Singapore, 138610, Republic of Singapore
| | - Helen Jingshu Jin
- Yale-NUS College, 12 College Ave West, #01- 201, Singapore, 138610, Republic of Singapore
| | - Jay B Lusk
- Yale-NUS College, 12 College Ave West, #01- 201, Singapore, 138610, Republic of Singapore
| | - Enrico Petretto
- Duke-NUS Medical School, 8 College Road, 169857, Singapore, Republic of Singapore
| | - Nicholas S Tolwinski
- Yale-NUS College, 12 College Ave West, #01- 201, Singapore, 138610, Republic of Singapore. .,Department of Biological Sciences, National University of Singapore, Block MD6, Centre for Translational Medicine, Yong Loo Lin School of Medicine, 14 Medical Drive, Level 10 South, 10-02M, Singapore, 117599, Republic of Singapore.
| |
Collapse
|
5
|
Otto GP, Sharma D, Williams RS. Non-Catalytic Roles of Presenilin Throughout Evolution. J Alzheimers Dis 2016; 52:1177-87. [PMID: 27079701 PMCID: PMC4927835 DOI: 10.3233/jad-150940] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2016] [Indexed: 12/20/2022]
Abstract
Research into Alzheimer's disease pathology and treatment has often focused on presenilin proteins. These proteins provide the key catalytic activity of the γ-secretase complex in the cleavage of amyloid-β precursor protein and resultant amyloid tangle deposition. Over the last 25 years, screening novel drugs to control this aberrant proteolytic activity has yet to identify effective treatments for the disease. In the search for other mechanisms of presenilin pathology, several studies have demonstrated that mammalian presenilin proteins also act in a non-proteolytic role as a scaffold to co-localize key signaling proteins. This role is likely to represent an ancestral presenilin function, as it has been described in genetically distant species including non-mammalian animals, plants, and a simple eukaryotic amoeba Dictyostelium that diverged from the human lineage over a billion years ago. Here, we review the non-catalytic scaffold role of presenilin, from mammalian models to other biomedical models, and include recent insights using Dictyostelium, to suggest that this role may provide an early evolutionary function of presenilin proteins.
Collapse
Affiliation(s)
- Grant P. Otto
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Devdutt Sharma
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Robin S.B. Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| |
Collapse
|
6
|
Duggan SP, McCarthy JV. Beyond γ-secretase activity: The multifunctional nature of presenilins in cell signalling pathways. Cell Signal 2015; 28:1-11. [PMID: 26498858 DOI: 10.1016/j.cellsig.2015.10.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/19/2015] [Indexed: 01/24/2023]
Abstract
The presenilins are the catalytic subunit of the membrane-embedded tetrameric γ-secretase protease complexes. More that 90 transmembrane proteins have been reported to be γ-secretase substrates, including the widely studied amyloid precursor protein (APP) and the Notch receptor, which are precursors for the generation of amyloid-β peptides and biologically active APP intracellular domain (AICD) and Notch intracellular domain (NICD). The diversity of γ-secretase substrates highlights the importance of presenilin-dependent γ-secretase protease activities as a regulatory mechanism in a range of biological systems. However, there is also a growing body of evidence that supports the existence of γ-secretase-independent functions for the presenilins in the regulation and progression of an array of cell signalling pathways. In this review, we will present an overview of current literature that proposes evolutionarily conserved presenilin functions outside of the γ-secretase complex, with a focus on the suggested role of the presenilins in the regulation of Wnt/β-catenin signalling, protein trafficking and degradation, calcium homeostasis and apoptosis.
Collapse
Affiliation(s)
- Stephen P Duggan
- Signal Transduction Laboratory, School of Biochemistry & Cell Biology, ABCRF, Western Gateway Building, University College Cork, Cork, Ireland
| | - Justin V McCarthy
- Signal Transduction Laboratory, School of Biochemistry & Cell Biology, ABCRF, Western Gateway Building, University College Cork, Cork, Ireland.
| |
Collapse
|
7
|
Zhang S, Zhang M, Cai F, Song W. Biological function of Presenilin and its role in AD pathogenesis. Transl Neurodegener 2013; 2:15. [PMID: 23866842 PMCID: PMC3718700 DOI: 10.1186/2047-9158-2-15] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 07/14/2013] [Indexed: 01/06/2023] Open
Abstract
Presenilins (PSs) are the catalytic core of γ-secretase complex. However, the mechanism of FAD-associated PS mutations in AD pathogenesis still remains elusive. Here we review the general biology and mechanism of γ-secretase and focus on the catalytic components – presenilins and their biological functions and contributions to the AD pathogenesis. The functions of presenilins are divided into γ-secretase dependent and γ-secretase independent ones. The γ-secretase dependent functions of presenilins are exemplified by the sequential cleavages in the processing of APP and Notch; the γ-secretase independent functions of presenilins include stabilizing β-catenin in Wnt signaling pathway, regulating calcium homeostasis and their interaction with synaptic transmission.
Collapse
Affiliation(s)
- Shuting Zhang
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| | | | | | | |
Collapse
|
8
|
Archbold HC, Yang YX, Chen L, Cadigan KM. How do they do Wnt they do?: regulation of transcription by the Wnt/β-catenin pathway. Acta Physiol (Oxf) 2012; 204:74-109. [PMID: 21624092 DOI: 10.1111/j.1748-1716.2011.02293.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Wnt/β-catenin signalling is known to play many roles in metazoan development and tissue homeostasis. Misregulation of the pathway has also been linked to many human diseases. In this review, specific aspects of the pathway's involvement in these processes are discussed, with an emphasis on how Wnt/β-catenin signalling regulates gene expression in a cell and temporally specific manner. The T-cell factor (TCF) family of transcription factors, which mediate a large portion of Wnt/β-catenin signalling, will be discussed in detail. Invertebrates contain a single TCF gene that contains two DNA-binding domains, the high mobility group (HMG) domain and the C-clamp, which increases the specificity of DNA binding. In vertebrates, the situation is more complex, with four TCF genes producing many isoforms that contain the HMG domain, but only some of which possess a C-clamp. Vertebrate TCFs have been reported to act in concert with many other transcription factors, which may explain how they obtain sufficient specificity for specific DNA sequences, as well as how they achieve a wide diversity of transcriptional outputs in different cells.
Collapse
Affiliation(s)
- H C Archbold
- Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, 48109-1048, USA
| | | | | | | |
Collapse
|
9
|
Zhou MN, Kunttas-Tatli E, Zimmerman S, Zhouzheng F, McCartney BM. Cortical localization of APC2 plays a role in actin organization but not in Wnt signaling in Drosophila. J Cell Sci 2011; 124:1589-600. [PMID: 21486956 DOI: 10.1242/jcs.073916] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The tumor suppressor Adenomatous polyposis coli (APC) has roles in both Wnt signaling and in actin and microtubule organization. Within the cell, APC proteins have been reported to localize in the cytoplasm, at the cell cortex and in the nucleus. How these localizations relate to the functions of the protein is an aspect of APC biology that is poorly understood. Using Drosophila S2 cells, we have dissected the structural and functional requirements for the cortical localization of Drosophila APC2. Here, we show that both the Armadillo repeats and a novel C-terminal domain are necessary for the cortical localization of APC2 in S2 cells and in the embryo, and that neither domain alone is sufficient for this localization. Furthermore, we show that the Armadillo repeats mediate self-association of APC2 molecules. To test the function of the cortical localization of APC2, we asked whether an APC2 protein deleted for the C-terminal localization domain could rescue APC mutant defects in Wnt signaling and actin organization in the Drosophila embryo. We show that although cortical localization is required for the APC2 function in organizing actin, cortical localization is dispensable for its role in regulating Wnt signaling.
Collapse
Affiliation(s)
- Meng-Ning Zhou
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
10
|
Harris ES, Nelson WJ. Adenomatous polyposis coli regulates endothelial cell migration independent of roles in beta-catenin signaling and cell-cell adhesion. Mol Biol Cell 2010; 21:2611-23. [PMID: 20519433 PMCID: PMC2912348 DOI: 10.1091/mbc.e10-03-0235] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 05/20/2010] [Accepted: 05/24/2010] [Indexed: 12/28/2022] Open
Abstract
Adenomatous polyposis coli (APC), a tumor suppressor commonly mutated in cancer, is a cytoskeletal organizer for cell migration and a scaffold for GSK3 beta/CKI-mediated phosphorylation and degradation of the Wnt effector beta-catenin. It remains unclear whether these different APC functions are coupled, or independently regulated and localized. In primary endothelial cells, we show that GSK3 beta/CKI-phosphorylated APC localizes to microtubule-dependent clusters at the tips of membrane extensions. Loss of GSK3 beta/CKI-phosphorylated APC from these clusters correlates with a decrease in cell migration. GSK3 beta/CKI-phosphorylated APC and beta-catenin at clusters is degraded rapidly by the proteasome, but inhibition of GSK3 beta/CKI does not increase beta-catenin-mediated transcription. GSK3 beta/CKI-phosphorylated and -nonphosphorylated APC also localize along adherens junctions, which requires actin and cell-cell adhesion. Significantly, inhibition of cell-cell adhesion results in loss of lateral membrane APC and a concomitant increase in GSK3 beta/CKI-phosphorylated APC in clusters. These results uncouple different APC functions and show that GSK3 beta/CKI phosphorylation regulates APC clusters and cell migration independently of cell-cell adhesion and beta-catenin transcriptional activity.
Collapse
Affiliation(s)
| | - W. James Nelson
- Departments of *Biology, and
- Molecular and Cellular Physiology, The James H. Clark Center, Bio-X Program, Stanford University, Stanford, CA 94305
| |
Collapse
|
11
|
Kessler T, Müller HAJ. Cleavage of Armadillo/beta-catenin by the caspase DrICE in Drosophila apoptotic epithelial cells. BMC DEVELOPMENTAL BIOLOGY 2009; 9:15. [PMID: 19232093 PMCID: PMC2657781 DOI: 10.1186/1471-213x-9-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Accepted: 02/20/2009] [Indexed: 11/24/2022]
Abstract
Background During apoptosis cells become profoundly restructured through concerted cleavage of cellular proteins by caspases. In epithelial tissues, apoptotic cells loose their apical/basal polarity and are extruded from the epithelium. We used the Drosophila embryo as a system to investigate the regulation of components of the zonula adherens during apoptosis. Since Armadillo/beta-catenin (Arm) is a major regulator of cadherin-mediated adhesion, we analyzed the mechanisms of Arm proteolysis in apoptosis. Results We define early and late apoptotic stages and find that early in apoptosis Dα-catenin remains relatively stable, while Arm and DE-cadherin protein levels are strongly reduced. Arm is cleaved by caspases in embryo extracts and we provide evidence that the caspase-3 homolog drICE cleaves Arm in vitro and in vivo. Cleavage by drICE creates a stable protein fragment that remains associated with the plasma membrane early in apoptosis. To further understand the role of caspase-mediated cleavage of Arm, we examined potential caspase cleavage sites and found that drICE cleaves Arm at a unique DQVD motif in the N-terminal domain of the protein. Mutation of the drICE cleavage site in Arm results in a protein that is not cleaved in vitro and in vivo. Furthermore we provide evidence that cleavage of Arm plays a role in the removal of DE-cadherin from the plasma membrane during apoptosis. Conclusion This study defines the specificity of caspase cleavage of Arm in Drosophila apoptotic cells. Our data suggest that N-terminal truncation of Arm by caspases is evolutionarily conserved and thus might provide a principal mechanism involved in the disassembly of adherens junctions during apoptosis.
Collapse
Affiliation(s)
- Thomas Kessler
- Institut für Genetik, Heinrich Heine Universität, Düsseldorf, Germany.
| | | |
Collapse
|
12
|
Raurell I, Codina M, Casagolda D, del Valle B, Baulida J, de Herreros AG, Duñach M. Gamma-secretase-dependent and -independent effects of presenilin1 on beta-catenin.Tcf-4 transcriptional activity. PLoS One 2008; 3:e4080. [PMID: 19114997 PMCID: PMC2603589 DOI: 10.1371/journal.pone.0004080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2008] [Accepted: 11/27/2008] [Indexed: 11/18/2022] Open
Abstract
Presenilin1 (PS1) is a component of the gamma-secretase complex mutated in cases of Familial Alzheimer's disease (FAD). PS1 is synthesized as a 50 kDa peptide subsequently processed to two 29 and 20 kDa subunits that remain associated. Processing of PS1 is inhibited by several mutations detected in FAD patients. PS1 acts as negative modulator of beta-catenin.Tcf-4 transcriptional activity. In this article we show that in murine embryonic fibroblasts (MEFs) the mechanisms of action of the processed and non-processed forms of PS1 on beta-catenin.Tcf-4 transcription are different. Whereas non-processed PS1 inhibits beta-catenin.Tcf-4 activity through a mechanism independent of gamma-secretase and associated with the interaction of this protein with plakoglobin and Tcf-4, the effect of processed PS1 is prevented by gamma-secretase inhibitors, and requires its interaction with E- or N-cadherin and the generation of cytosolic terminal fragments of these two cadherins, which in turn destabilize the beta-catenin transcriptional cofactor CBP. Accordingly, the two forms of PS1 interact differently with E-cadherin or beta-catenin and plakoglobin: whereas processed PS1 binds E-cadherin with high affinity and beta-catenin or plakoglobin weakly, the non-processed form behaves inversely. Moreover, contrarily to processed PS1, that decreases the levels of c-fos RNA, non-processed PS1 inhibits the expression c-myc, a known target of beta-catenin.Tcf-4, and does not block the activity of other transcriptional factors requiring CBP. These results indicate that prevention of PS1 processing in FAD affects the mechanism of repression of the transcriptional activity dependent on beta-catenin.
Collapse
Affiliation(s)
- Imma Raurell
- Unitat de Biofísica-CEB, Departament de Bioquímica i Biologia Molecular, Facultat Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Montserrat Codina
- Unitat de Biofísica-CEB, Departament de Bioquímica i Biologia Molecular, Facultat Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Programa de Recerca en Càncer, IMIM-Hospital del Mar, Barcelona, Spain
| | - David Casagolda
- Unitat de Biofísica-CEB, Departament de Bioquímica i Biologia Molecular, Facultat Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Beatriz del Valle
- Unitat de Biofísica-CEB, Departament de Bioquímica i Biologia Molecular, Facultat Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josep Baulida
- Programa de Recerca en Càncer, IMIM-Hospital del Mar, Barcelona, Spain
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, IMIM-Hospital del Mar, Barcelona, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
- * E-mail: (MD); (AGdH)
| | - Mireia Duñach
- Unitat de Biofísica-CEB, Departament de Bioquímica i Biologia Molecular, Facultat Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- * E-mail: (MD); (AGdH)
| |
Collapse
|
13
|
Hass MR, Sato C, Kopan R, Zhao G. Presenilin: RIP and beyond. Semin Cell Dev Biol 2008; 20:201-10. [PMID: 19073272 DOI: 10.1016/j.semcdb.2008.11.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/19/2008] [Accepted: 11/19/2008] [Indexed: 12/22/2022]
Abstract
Over the years the presenilins (PSENs), a family of multi-transmembrane domain proteins, have been ascribed a number of diverse potential functions. Recent in vivo evidence has supported the existence of PSEN functions beyond its well-established role in regulated intramembrane proteolysis. In this review, we will briefly discuss the ability of PSEN to modulate cellular signaling pathways through gamma-secretase cleavage of transmembrane proteins. Additionally, we will critically examine the proposed roles of PSEN in the regulation of beta-catenin function, protein trafficking, calcium regulation, and apoptosis.
Collapse
Affiliation(s)
- Matthew R Hass
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, United States
| | | | | | | |
Collapse
|
14
|
Fiehler RW, Wolff T. Drosophila Myosin II, Zipper, is essential for ommatidial rotation. Dev Biol 2007; 310:348-62. [PMID: 17826761 PMCID: PMC2110880 DOI: 10.1016/j.ydbio.2007.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 06/20/2007] [Accepted: 08/01/2007] [Indexed: 01/17/2023]
Abstract
The adult Drosophila retina is a highly polarized epithelium derived from a precursor tissue that is initially symmetric across its dorsoventral axis. Specialized 90 degrees rotational movements of subsets of cells, the ommatidial precursors, establish mirror symmetry in the retinal epithelium. Myosin II, or Zipper (Zip), a motor protein, regulates the rate at which ommatidia rotate: in zip mutants, the rate of rotation is significantly slowed. Zip is concentrated in the cells that we show to be at the likely interface between rotating and non-rotating cells: the boundary between differentiated and undifferentiated cells. Zip is also robust in newly added ommatidial cells, consistent with our model that the machinery that drives rotation should shift to newly recruited cells as they are added to the growing ommatidium. Finally, cell death genes and canonical Wnt signaling pathway members genetically modify the zip phenotype.
Collapse
Affiliation(s)
- Ryan W Fiehler
- Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
15
|
VanHook A, Letsou A. Head involution inDrosophila: Genetic and morphogenetic connections to dorsal closure. Dev Dyn 2007; 237:28-38. [DOI: 10.1002/dvdy.21405] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
16
|
Luo X, Puig O, Hyun J, Bohmann D, Jasper H. Foxo and Fos regulate the decision between cell death and survival in response to UV irradiation. EMBO J 2006; 26:380-90. [PMID: 17183370 PMCID: PMC1783446 DOI: 10.1038/sj.emboj.7601484] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Accepted: 11/08/2006] [Indexed: 01/23/2023] Open
Abstract
Cells damaged by environmental insults have to be repaired or eliminated to ensure tissue homeostasis in metazoans. Recent studies suggest that the balance between cell survival signals and pro-apoptotic stimuli controls the decision between cell repair and death. How these competing signals are integrated and interpreted to achieve accurate control over cell fate in vivo is incompletely understood. Here, we show that the Forkhead Box O transcription factor Foxo and the AP-1 transcription factor DFos are required downstream of Jun-N-terminal kinase signaling for the apoptotic response to UV-induced DNA damage in the developing Drosophila retina. Both transcription factors regulate the pro-apoptotic gene hid. Our results indicate that UV-induced apoptosis is repressed by receptor tyrosine kinase-mediated inactivation of Foxo. These data suggest that integrating stress and survival signals through Foxo drives the decision between cell death and repair of damaged cells in vivo.
Collapse
Affiliation(s)
- Xi Luo
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Oscar Puig
- Institute of Biotechnology, University of Helsinki, Viikinkaari, Finland
| | - Joogyung Hyun
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Dirk Bohmann
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Heinrich Jasper
- Department of Biology, University of Rochester, Rochester, NY, USA
- Department of Biology, University of Rochester, River Campus Box 270211, Rochester, NY 14627, USA. Tel.: +1 585 275 8973; Fax: +1 585 275 2070; E-mail:
| |
Collapse
|
17
|
Price MH, Roberts DM, McCartney BM, Jezuit E, Peifer M. Cytoskeletal dynamics and cell signaling during planar polarity establishment in theDrosophilaembryonic denticle. J Cell Sci 2006; 119:403-15. [PMID: 16418222 DOI: 10.1242/jcs.02761] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many epithelial cells are polarized along the plane of the epithelium, a property termed planar cell polarity. The Drosophila wing and eye imaginal discs are the premier models of this process. Many proteins required for polarity establishment and its translation into cytoskeletal polarity were identified from studies of those tissues. More recently, several vertebrate tissues have been shown to exhibit planar cell polarity. Striking similarities and differences have been observed when different tissues exhibiting planar cell polarity are compared. Here we describe a new tissue exhibiting planar cell polarity – the denticles, hair-like projections of the Drosophila embryonic epidermis. We describe in real time the changes in the actin cytoskeleton that underlie denticle development, and compare this with the localization of microtubules, revealing new aspects of cytoskeletal dynamics that may have more general applicability. We present an initial characterization of the localization of several actin regulators during denticle development. We find that several core planar cell polarity proteins are asymmetrically localized during the process. Finally, we define roles for the canonical Wingless and Hedgehog pathways and for core planar cell polarity proteins in denticle polarity.
Collapse
Affiliation(s)
- Meredith H Price
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | |
Collapse
|
18
|
Raurell I, Castaño J, Francí C, García de Herreros A, Duñach M. Presenilin-1 interacts with plakoglobin and enhances plakoglobin-Tcf-4 association. Implications for the regulation of beta-catenin/Tcf-4-dependent transcription. J Biol Chem 2005; 281:1401-11. [PMID: 16306047 DOI: 10.1074/jbc.m508153200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alzheimer disease-linked Presenilin-1 (PS1) is a negative modulator of beta-catenin/Tcf-4 activity. However, the mechanism underlying this effect is not well understood. We show here that the effects of PS1 on the activity of this complex in epithelial cells are independent of its gamma-secretase activity and its interaction with beta-catenin. As presented in this report PS1 also binds plakoglobin with similar affinity as beta-catenin, although this interaction does not involve equivalent residues in the two catenins. Moreover, PS1 association with plakoglobin enhances the interaction of this molecule with Tcf-4 and prevents its binding to DNA. These effects were observed with the unprocessed form of PS1, which has higher affinity for plakoglobin and beta-catenin than processed PS1. These results provide a new explanation for the effects of PS1 on gene transcription mediated by beta-catenin in epithelial cells.
Collapse
Affiliation(s)
- Imma Raurell
- Unitat de Biofísica, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | | | | | | | | |
Collapse
|
19
|
Werz C, Lee TV, Lee PL, Lackey M, Bolduc C, Stein DS, Bergmann A. Mis-specified cells die by an active gene-directed process, and inhibition of this death results in cell fate transformation in Drosophila. Development 2005; 132:5343-52. [PMID: 16280349 PMCID: PMC2760325 DOI: 10.1242/dev.02150] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Incorrectly specified or mis-specified cells often undergo cell death or are transformed to adopt a different cell fate during development. The underlying cause for this distinction is largely unknown. In many developmental mutants in Drosophila, large numbers of mis-specified cells die synchronously, providing a convenient model for analysis of this phenomenon. The maternal mutant bicoid is particularly useful model with which to address this issue because its mutant phenotype is a combination of both transformation of tissue (acron to telson) and cell death in the presumptive head and thorax regions. We show that a subset of these mis-specified cells die through an active gene-directed process involving transcriptional upregulation of the cell death inducer hid. Upregulation of hid also occurs in oskar mutants and other segmentation mutants. In hid bicoid double mutants, mis-specified cells in the presumptive head and thorax survive and continue to develop, but they are transformed to adopt a different cell fate. We provide evidence that the terminal torso signaling pathway protects the mis-specified telson tissue in bicoid mutants from hid-induced cell death, whereas mis-specified cells in the head and thorax die, presumably because equivalent survival signals are lacking. These data support a model whereby mis-specification can be tolerated if a survival pathway is provided, resulting in cellular transformation.
Collapse
Affiliation(s)
- Christian Werz
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
| | - Tom V. Lee
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
- The Genes and Development Graduate Program (http://www.mdanderson.org/genedev)
| | - Peter L. Lee
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
| | - Melinda Lackey
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
| | - Clare Bolduc
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
| | - David S. Stein
- The University of Texas at Austin, Patterson labs 532, Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, 2401 W24th and Speedway, Austin, TX 78712, USA
| | - Andreas Bergmann
- The University of Texas M.D. Anderson Cancer Center, Department of Biochemistry and Molecular Biology, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
- The Genes and Development Graduate Program (http://www.mdanderson.org/genedev)
- Author for correspondence (e-mail: )
| |
Collapse
|
20
|
Marambaud P, Robakis NK. Genetic and molecular aspects of Alzheimer's disease shed light on new mechanisms of transcriptional regulation. GENES BRAIN AND BEHAVIOR 2005; 4:134-46. [PMID: 15810902 DOI: 10.1111/j.1601-183x.2005.00086.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Rapid advances made in biological research aimed at understanding the molecular basis of the pathogenesis of Alzheimer's disease have led to the characterization of a novel catalytic activity termed gamma-secretase. First described for its beta-amyloid-producing function, gamma-secretase is now actively studied for its role in a novel signal transduction paradigm, which implicates cell-surface receptor proteolysis and direct surface-to-nucleus signal transduction. gamma-Secretase targets numerous type I protein receptors involved in diverse functions ranging from normal development to neurodegeneration. In this Review we discuss how the study of the genetic and molecular aspects of Alzheimer's disease has revealed a dual role of gamma-secretase in transcriptional regulation and in the pathogenesis of familial Alzheimer's disease.
Collapse
Affiliation(s)
- P Marambaud
- Department of Psychiatry and Fishberg Research Center for Neurobiology, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | |
Collapse
|
21
|
Desbordes SC, Chandraratna D, Sanson B. A screen for genes regulating the wingless gradient in Drosophila embryos. Genetics 2005; 170:749-66. [PMID: 15834138 PMCID: PMC1450427 DOI: 10.1534/genetics.105.040667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During the development of the Drosophila embryonic epidermis, the secreted Wingless protein initially spreads symmetrically from its source. At later stages, Wingless becomes asymmetrically distributed in a Hedgehog-dependent manner, to control the patterning of the embryonic epidermis. When Wingless is misexpressed in engrailed cells in hedgehog heterozygous mutant embryos, larvae show a dominant phenotype consisting of patches of naked cuticle in denticle belts. This dose-sensitive phenotype is a direct consequence of a change in Wg protein distribution. We used this phenotype to carry out a screen for identifying genes regulating Wingless distribution or transport in the embryonic epidermis. Using a third chromosome deficiency collection, we found several genomic regions that showed a dominant interaction. After using a secondary screen to test for mutants and smaller deficiencies, we identified three interacting genes: dally, notum, and brahma. We confirmed that dally, as well as its homolog dally-like, and notum affect Wingless distribution in the embryonic epidermis, directly or indirectly. Thus, our assay can be used effectively to screen for genes regulating Wingless distribution or transport.
Collapse
|
22
|
Yeo W, Gautier J. Early neural cell death: dying to become neurons. Dev Biol 2004; 274:233-44. [PMID: 15385155 DOI: 10.1016/j.ydbio.2004.07.026] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Revised: 07/23/2004] [Accepted: 07/28/2004] [Indexed: 10/26/2022]
Abstract
The importance of programmed cell death (PCD) during vertebrate development has been well established. During the development of the nervous system in particular, neurotrophic cell death in innervating neurons matches the number of neurons to the size of their target field. However, PCD also occurs during earlier stages of neural development, within populations of proliferating neural precursors and newly postmitotic neuroblasts, all of which are not yet fully differentiated. This review addresses early neural PCD, which is distinct from neurotrophic death in differentiated neurons. Although early neural PCD is observed in a range of organisms, from Caenorhabditis elegans to mouse, the role and the regulation of early neural PCD are not well understood. The regulation of early neural PCD can be inferred from the function of factors such as bone morphogenetic proteins (BMPs), Wnts, fibroblast growth factors (FGFs), and Sonic Hedgehog (Shh), which regulate both early neural development and PCD occurring in other developmental processes. Cell number control, removal of damaged or misspecified cells (spatially or temporally), and selection are the proposed roles early neural PCDs play during neural development. Data from developmental PCD in C. elegans and Drosophila provide insights into the possible signaling pathways integrating PCD with other processes during early neural development and the roles they might play.
Collapse
Affiliation(s)
- Weeteck Yeo
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
23
|
Dermaut B, Kumar-Singh S, Engelborghs S, Theuns J, Rademakers R, Saerens J, Pickut BA, Peeters K, van den Broeck M, Vennekens K, Claes S, Cruts M, Cras P, Martin JJ, Van Broeckhoven C, De Deyn PP. A novel presenilin 1 mutation associated with Pick's disease but not beta-amyloid plaques. Ann Neurol 2004; 55:617-26. [PMID: 15122701 DOI: 10.1002/ana.20083] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Familial forms of frontotemporal dementia (FTD) with tauopathy are mostly caused by mutations in the gene encoding the microtubule-associated protein tau (MAPT). However, rare forms of familial tauopathy without MAPT mutations have been reported, suggesting other tauopathy-related genetic defects. Interestingly, two presenilin 1 (PS1) mutations (Leu113Pro and insArg352) recently have been associated with familial FTD albeit without neuropathological confirmation. We report here a novel PS1 mutation in a patient with Pick-type tauopathy in the absence of extracellular beta-amyloid deposits. The mutation is predicted to substitute Gly-->Val at codon position 183 (Gly183Val) and to affect the splice signal at the junction of the sixth exon and intron. Further clinical-genetic investigation showed a positive family history of FTD-like dementia and suggested that Gly183Val is associated with a phenotypically heterogeneous neurodegenerative disorder. Our results suggest PS1 as a candidate gene for Pick-type tauopathy without MAPT mutations.
Collapse
Affiliation(s)
- Bart Dermaut
- Department of Molecular Genetics, Flanders Interuniversity Institute of Biotechnology (VIB8), University of Antwerp, Antwerpen, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Johnston LA, Sanders AL. Wingless promotes cell survival but constrains growth during Drosophila wing development. Nat Cell Biol 2003; 5:827-33. [PMID: 12942089 DOI: 10.1038/ncb1041] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Accepted: 08/05/2003] [Indexed: 11/09/2022]
Abstract
During animal development, organs grow to a fixed size and shape. Organ development typically begins with a rapid growth phase followed by a gradual decline in growth rate as the organ matures, but the regulation of either stage of growth remains unclear. The Wnt/Wingless (Wg) proteins are critical for patterning most animal organs, have diverse effects on development and have been proposed to promote organ growth. Here we report that contrary to this view, Wg activity actually constrains wing growth during Drosophila melanogaster wing development. In addition, we demonstrate that Wg is required for wing cell survival, particularly during the rapid growth phase of wing development. We propose that the cell-survival- and growth-constraining activities of Wg function to sculpt and delimit final wing size as part of its overall patterning programme.
Collapse
Affiliation(s)
- Laura A Johnston
- Department of Genetics & Development, College of Physicians & Surgeons Columbia University 701 West 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
25
|
McDonald JA, Pinheiro EM, Montell DJ. PVF1, a PDGF/VEGF homolog, is sufficient to guide border cells and interacts genetically with Taiman. Development 2003; 130:3469-78. [PMID: 12810594 DOI: 10.1242/dev.00574] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The border cells of the Drosophila ovary undergo a well-defined and developmentally regulated cell migration. Two signals have previously been shown to control where and when the cells migrate. The steroid hormone ecdysone, acting through its receptor and a coactivator known as Taiman, contributes to regulating the timing of border cell migration. PVF1, a growth factor related to platelet-derived growth factor and vascular-endothelial growth factor, contributes to guiding the border cells to the oocyte. To probe the mechanisms controlling border cell migration further, we performed a screen for genes that exhibit dominant genetic interactions with taiman. We identified 14 genomic regions that interact with taiman. Within one region, we identified Pvf1 as the gene responsible for the interaction. Signaling by PVF1 has been proposed to guide the border cells to their proper target, but ectopic PVF1 has not been tested for its ability to redirect the border cells. We tested the ability of PVF1, as well as other factors such as Gurken, to guide the border cells to new targets. Our results demonstrate that ectopic expression of PVF1 is sufficient to redirect border cells in some egg chambers but that the other factors tested are not. These data suggest that the guidance of border cell migration is robust and that there are likely to be additional factors that contribute to long-range guidance of these cells. In addition, we find that taiman and Pvf1 regulate the dynamic localization of E-cadherin in the border cells, possibly accounting for the interaction between these two pathways.
Collapse
Affiliation(s)
- Jocelyn A McDonald
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205-2185, USA
| | | | | |
Collapse
|
26
|
Albanese C, Wu K, D'Amico M, Jarrett C, Joyce D, Hughes J, Hulit J, Sakamaki T, Fu M, Ben-Ze'ev A, Bromberg JF, Lamberti C, Verma U, Gaynor RB, Byers SW, Pestell RG. IKKalpha regulates mitogenic signaling through transcriptional induction of cyclin D1 via Tcf. Mol Biol Cell 2003; 14:585-99. [PMID: 12589056 PMCID: PMC149994 DOI: 10.1091/mbc.02-06-0101] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The Wnt/beta-catenin/Tcf and IkappaB/NF-kappaB cascades are independent pathways involved in cell cycle control, cellular differentiation, and inflammation. Constitutive Wnt/beta-catenin signaling occurs in certain cancers from mutation of components of the pathway and from activating growth factor receptors, including RON and MET. The resulting accumulation of cytoplasmic and nuclear beta-catenin interacts with the Tcf/LEF transcription factors to induce target genes. The IkappaB kinase complex (IKK) that phosphorylates IkappaB contains IKKalpha, IKKbeta, and IKKgamma. Here we show that the cyclin D1 gene functions as a point of convergence between the Wnt/beta-catenin and IkappaB pathways in mitogenic signaling. Mitogenic induction of G(1)-S phase progression and cyclin D1 expression was PI3K dependent, and cyclin D1(-/-) cells showed reduced PI3K-dependent S-phase entry. PI3K-dependent induction of cyclin D1 was blocked by inhibitors of PI3K/Akt/IkappaB/IKKalpha or beta-catenin signaling. A single Tcf site in the cyclin D1 promoter was required for induction by PI3K or IKKalpha. In IKKalpha(-/-) cells, mitogen-induced DNA synthesis, and expression of Tcf-responsive genes was reduced. Reintroduction of IKKalpha restored normal mitogen induction of cyclin D1 through a Tcf site. In IKKalpha(-/-) cells, beta-catenin phosphorylation was decreased and purified IKKalpha was sufficient for phosphorylation of beta-catenin through its N-terminus in vitro. Because IKKalpha but not IKKbeta induced cyclin D1 expression through Tcf activity, these studies indicate that the relative levels of IKKalpha and IKKbeta may alter their substrate and signaling specificities to regulate mitogen-induced DNA synthesis through distinct mechanisms.
Collapse
Affiliation(s)
- Chris Albanese
- The Albert Einstein Cancer Center, Division of Hormone-Dependent Tumor Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Meredith JE, Wang Q, Mitchell TJ, Olson RE, Zaczek R, Stern AM, Seiffert D. Gamma-secretase activity is not involved in presenilin-mediated regulation of beta-catenin. Biochem Biophys Res Commun 2002; 299:744-50. [PMID: 12470641 DOI: 10.1016/s0006-291x(02)02747-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Presenilins (PS) are involved in gamma-secretase-mediated processing of beta-amyloid precursor protein (APP) and the Notch family of proteins. In addition, presenilin 1 (PS-1) binds to members of the armadillo family of proteins. In this study the relationship between PS-1-mediated proteolytic activity and PS-1-mediated regulation of beta-catenin function was investigated. Incubation of cells with a potent, small molecule gamma-secretase inhibitor did not affect PS-1/beta-catenin interaction as determined by co-immunoprecipitation, or affect the regulation of beta-catenin turnover, as determined by pulse-chase analysis, even at inhibitor concentrations that completely blocked PS-mediated APP processing. Moreover, inhibition of PS-1-mediated proteolytic activity did not affect beta-catenin trafficking, as determined by immunolocalization and immunoblotting, or beta-catenin-mediated transcription. These results indicate that PS-1-mediated regulation of gamma-secretase activity and PS-1-mediated regulation of beta-catenin function can be pharmacologically separated and support the idea that these are distinct functions.
Collapse
Affiliation(s)
- Jere E Meredith
- Bristol-Myers Squibb Company, The Pharmaceutical Research Institute, Experimental Station, Route 141 and Henry Clay Rd., Wilmington, DE 19880, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Mirkovic I, Charish K, Gorski SM, McKnight K, Verheyen EM. Drosophila nemo is an essential gene involved in the regulation of programmed cell death. Mech Dev 2002; 119:9-20. [PMID: 12385750 DOI: 10.1016/s0925-4773(02)00289-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nemo-like kinases define a novel family of serine/threonine kinases that are involved in integrating multiple signaling pathways. They are conserved regulators of Wnt/Wingless pathways, which may coordinate Wnt with TGFbeta-mediated signaling. Drosophila nemo was identified through its involvement in epithelial planar polarity, a process regulated by a non-canonical Wnt pathway. We have previously found that ectopic expression of Nemo using the Gal4-UAS system resulted in embryonic lethality associated with defects in patterning and head development. In this study we present our analyses of the phenotypes of germline clone-derived embryos. We observe lethality associated with head defects and reduction of programmed cell death and conclude that nmo is an essential gene. We also present data showing that nmo is involved in regulating apoptosis during eye development, based on both loss of function phenotypes and on genetic interactions with the pro-apoptotic gene reaper. Finally, we present genetic data from the adult wing that suggest the activity of ectopically expressed Nemo can be modulated by Jun N-terminal kinase (JNK) signaling. Such an observation supports the model that there is cross-talk between Wnt, TGFbeta and JNK signaling at multiple stages of development.
Collapse
Affiliation(s)
- Ivana Mirkovic
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | | | | | | | | |
Collapse
|
29
|
Kang DE, Soriano S, Xia X, Eberhart CG, De Strooper B, Zheng H, Koo EH. Presenilin couples the paired phosphorylation of beta-catenin independent of axin: implications for beta-catenin activation in tumorigenesis. Cell 2002; 110:751-62. [PMID: 12297048 DOI: 10.1016/s0092-8674(02)00970-4] [Citation(s) in RCA: 191] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The Alzheimer's disease-linked gene presenilin 1 (PS1) is required for intramembrane proteolysis of APP and Notch. In addition, recent observations strongly implicate PS1 as a negative regulator of the Wnt/beta-catenin signaling pathway, although the mechanism underlying this activity is unknown. Here, we show that presenilin functions as a scaffold that rapidly couples beta-catenin phosphorylation through two sequential kinase activities independent of the Wnt-regulated Axin/CK1alpha complex. Thus, presenilin deficiency results in increased beta-catenin stability in vitro and in vivo by disconnecting the stepwise phosphorylation of beta-catenin, both in the presence and absence of Wnt stimulation. These findings highlight an aspect of beta-catenin regulation outside of the canonical Wnt-regulated pathway and a function of presenilin separate from intramembrane proteolysis.
Collapse
Affiliation(s)
- David E Kang
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Galis F, van Dooren TJM, Metz JAJ. Conservation of the segmented germband stage: robustness or pleiotropy? Trends Genet 2002; 18:504-9. [PMID: 12350339 DOI: 10.1016/s0168-9525(02)02739-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gene expression patterns of the segment polarity genes in the extended and segmented germband stage are remarkably conserved among insects. To explain the conservation of these stages, two hypotheses have been proposed. One hypothesis states that the conservation reflects a high interactivity between modules, so that mutations would have several pleiotropic effects in other parts of the body, resulting in stabilizing selection against mutational variation. The other hypothesis states that the conservation is caused by robustness of the segment polarity network against mutational changes. When evaluating the empirical evidence for these hypotheses, we found strong support for pleiotropy and little evidence supporting robustness of the segment polarity network. This points to a key role for stabilizing selection in the conservation of these stages. Finally, we discuss the implications for robustness of organizers and long-term conservation in general.
Collapse
Affiliation(s)
- Frietson Galis
- Institute of Evolutionary and Ecological Sciences, Leiden University, PO Box 9516, 2300RA Leiden University, The Netherlands.
| | | | | |
Collapse
|
31
|
You Z, Saims D, Chen S, Zhang Z, Guttridge DC, Guan KL, MacDougald OA, Brown AMC, Evan G, Kitajewski J, Wang CY. Wnt signaling promotes oncogenic transformation by inhibiting c-Myc-induced apoptosis. J Cell Biol 2002; 157:429-40. [PMID: 11980918 PMCID: PMC2173296 DOI: 10.1083/jcb.200201110] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aberrant activation of the Wnt/beta-catenin signaling pathway is associated with numerous human cancers and often correlates with the overexpression or amplification of the c-myc oncogene. Paradoxical to the cellular transformation potential of c-Myc is its ability to also induce apoptosis. Using an inducible c-MycER expression system, we found that Wnt/beta-catenin signaling suppressed apoptosis by inhibiting c-Myc-induced release of cytochrome c and caspase activation. Both cyclooxygenase 2 and WISP-1 were identified as effectors of the Wnt-mediated antiapoptotic signal. Soft agar assays showed that neither c-Myc nor Wnt-1 alone was sufficient to induce cellular transformation, but that Wnt and c-Myc coordinated in inducing transformation. Furthermore, coexpression of Wnt-1 and c-Myc induced high-frequency and rapid tumor growth in nude mice. Extensive apoptotic bodies were characteristic of c-Myc-induced tumors, but not tumors induced by coactivation of c-Myc and Wnt-1, indicating that the antiapoptotic function of Wnt-1 plays a critical role in the synergetic action between c-Myc and Wnt-1. These results elucidate the molecular mechanisms by which Wnt/beta-catenin inhibits apoptosis and provide new insight into Wnt signaling-mediated oncogenesis.
Collapse
Affiliation(s)
- Zongbing You
- Laboratory of Molecular Signaling and Apoptosis, Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor 48109-1078, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Killick R, Pollard CC, Asuni AA, Mudher AK, Richardson JC, Rupniak HT, Sheppard PW, Varndell IM, Brion JP, Levey AI, Levy OA, Vestling M, Cowburn R, Lovestone S, Anderton BH. Presenilin 1 independently regulates beta-catenin stability and transcriptional activity. J Biol Chem 2001; 276:48554-61. [PMID: 11606587 DOI: 10.1074/jbc.m108332200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Presenilin 1 (PS1) regulates beta-catenin stability; however, published data regarding the direction of the effect are contradictory. We examined the effects of wild-type and mutant forms of PS1 on the membrane, cytoplasmic, nuclear, and signaling pools of endogenous and exogenous beta-catenin by immunofluorescence microscopy, subcellular fractionation, and in a transcription assay. We found that PS1 destabilizes the cytoplasmic and nuclear pools of beta-catenin when stabilized by Wnt or Dvl but not when stabilized at lower levels of the Wnt pathway. The PS1 mutants examined were less able to reduce the stability of beta-catenin. PS1 also inhibited the transcriptional activity of endogenous beta-catenin, and the PS1 mutants were again less inhibitory at the level of Dvl but showed a different pattern of inhibition toward transcription below Dvl. The transcriptional activity of exogenously expressed wild-type beta-catenin and two mutants, DeltaN89beta-catenin and DeltaSTbeta-catenin, were also inhibited by wild-type and mutant PS1. We conclude that PS1 negatively regulates the stability and transcriptional activity of beta-catenin at different levels in the Wnt pathway, that the effect on transcriptional activity appears to be independent of the GSK-3beta mediated degradation of beta-catenin, and that mutations in PS1 differentially affect the stability and transcriptional activity of beta-catenin.
Collapse
Affiliation(s)
- R Killick
- Department of Neuroscience, Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London SE5 8AF, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xia X, Qian S, Soriano S, Wu Y, Fletcher AM, Wang XJ, Koo EH, Wu X, Zheng H. Loss of presenilin 1 is associated with enhanced beta-catenin signaling and skin tumorigenesis. Proc Natl Acad Sci U S A 2001; 98:10863-8. [PMID: 11517342 PMCID: PMC58565 DOI: 10.1073/pnas.191284198] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2001] [Indexed: 01/08/2023] Open
Abstract
Presenilin 1 (PS1) is required for the proteolytic processing of Notch and the beta-amyloid precursor protein (APP), molecules that play pivotal roles in cell-fate determination during development and Alzheimer's disease pathogenesis, respectively. In addition, PS1 interacts with beta-catenin and promotes its turnover through independent mechanisms. Consistent with this activity, we report here that PS1 is important in controlling epidermal cell proliferation in vivo. PS1 knockout mice that are rescued through neuronal expression of human PS1 transgene develop spontaneous skin cancers. PS1-null keratinocytes exhibit higher cytosolic beta-catenin and beta-catenin/lymphoid enhancer factor-1/T cell factor (beta-catenin/LEF)-mediated signaling. This effect can be reversed by reintroducing wild-type PS1, but not a PS1 mutant active in Notch processing but defective in beta-catenin binding. Nuclear beta-catenin protein can be detected in tumors. Elevated beta-catenin/LEF signaling is correlated with activation of its downstream target cyclin D1 and accelerated entry from G(1) into S phase of the cell cycle. This report demonstrates a function of PS1 in adult tissues, and our analysis suggests that deregulation of beta-catenin pathway contributes to the skin tumor phenotype.
Collapse
Affiliation(s)
- X Xia
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- B De Strooper
- Center for Human Genetics, Neuronal Cell Biology Laboratory, Katholieke Universiteit Leuven and Flanders Interuniversitary Institute for Biotechnology, 3000 Leuven, Belgium.
| | | |
Collapse
|
35
|
Soriano S, Kang DE, Fu M, Pestell R, Chevallier N, Zheng H, Koo EH. Presenilin 1 negatively regulates beta-catenin/T cell factor/lymphoid enhancer factor-1 signaling independently of beta-amyloid precursor protein and notch processing. J Cell Biol 2001; 152:785-94. [PMID: 11266469 PMCID: PMC2195782 DOI: 10.1083/jcb.152.4.785] [Citation(s) in RCA: 173] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2000] [Accepted: 01/05/2001] [Indexed: 01/07/2023] Open
Abstract
In addition to its documented role in the proteolytic processing of Notch-1 and the beta-amyloid precursor protein, presenilin 1 (PS1) associates with beta-catenin. In this study, we show that this interaction plays a critical role in regulating beta-catenin/T Cell Factor/Lymphoid Enhancer Factor-1 (LEF) signaling. PS1 deficiency results in accumulation of cytosolic beta-catenin, leading to a beta-catenin/LEF-dependent increase in cyclin D1 transcription and accelerated entry into the S phase of the cell cycle. Conversely, PS1 specifically represses LEF-dependent transcription in a dose-dependent manner. The hyperproliferative response can be reversed by reintroducing PS1 expression or overexpressing axin, but not a PS1 mutant that does not bind beta-catenin (PS1Deltacat) or by two different familial Alzheimer's disease mutants. In contrast, PS1Deltacat restores Notch-1 proteolytic cleavage and Abeta generation in PS1-deficient cells, indicating that PS1 function in modulating beta-catenin levels can be separated from its roles in facilitating gamma-secretase cleavage of beta-amyloid precursor protein and in Notch-1 signaling. Finally, we show an altered response to Wnt signaling and impaired ubiquitination of beta-catenin in the absence of PS1, a phenotype that may account for the increased stability in PS1-deficient cells. Thus, PS1 adds to the molecules that are known to regulate the rapid turnover of beta-catenin.
Collapse
Affiliation(s)
- Salvador Soriano
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - David E. Kang
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Maofu Fu
- The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Richard Pestell
- The Albert Einstein Cancer Center, Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Nathalie Chevallier
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Hui Zheng
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Edward H. Koo
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
36
|
Noll E, Medina M, Hartley D, Zhou J, Perrimon N, Kosik KS. Presenilin affects arm/beta-catenin localization and function in Drosophila. Dev Biol 2000; 227:450-64. [PMID: 11071766 DOI: 10.1006/dbio.2000.9925] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Presenilin is an essential gene for development that when disrupted leads to a neurogenic phenotype that closely resembles Notch loss of function in Drosophila. In humans, many naturally occurring mutations in Presenilin 1 or 2 cause early onset Alzheimer's disease. Both loss of expression and overexpression of Presenilin suggested a role for this protein in the localization of Armadillo/beta-catenin. In blastoderm stage Presenilin mutants, Arm is aberrantly distributed, often in Ubiquitin-immunoreactive cytoplasmic inclusions predominantly located basally in the cell. These inclusions were not observed in loss of function Notch mutants, suggesting that failure to process Notch is not the only consequence of the loss of Presenilin function. Human presenilin 1 expressed in Drosophila produces embryonic phenotypes resembling those associated with mutations in Armadillo and exhibited reduced Armadillo at the plasma membrane that is likely due to retention of Armadillo in a complex with Presenilin. The interaction between Armadillo/beta-catenin and Presenilin 1 requires a third protein which may be delta-catenin. Our results suggest that Presenilin may regulate the delivery of a multiprotein complex that regulates Armadillo trafficking between the adherens junction and the proteasome.
Collapse
Affiliation(s)
- E Noll
- Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|