1
|
Elliott C, Jackson J, Findlay J, Williams G, Ghosh A, Ribe E, Ulmschneider M, Khan A, Ballard C, Aarsland D, Baillie GS, Harte M, Killick R. Blocking the Dkk1-LRP6 interaction prevents acute amyloid-β-driven cognitive impairment. Cell Signal 2025; 131:111716. [PMID: 40057150 DOI: 10.1016/j.cellsig.2025.111716] [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: 01/24/2025] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025]
Abstract
Synapse loss driven by amyloid-β (Aβ) is an early event in Alzheimer's disease (AD). Although the mechanism by which Aβ drives synapse loss remain poorly understood data indicate that a disruption of Wnt signalling plays an important part. We have shown that Aβ exerts its effects on synapses through Dickkopf-1 (Dkk1), a secreted protein that acts upon Wnt signalling via a direct interaction with the canonical Wnt pathway co-receptor proteins, LRP5 and LRP6, preventing their interaction with the receptor Frizzled. This antagonises canonical, Wnt/β-catenin, signalling and allows concomitant activation of non-canonical signalling pathways. We contend that it is the switch from canonical to non-canonical Wnt signalling activity that drives synapse loss and subsequent cognitive impairment in AD, driven by Aβ and mediated by Dkk1. Preventing the Dkk1-LRP5/6 interaction could protect synapses and cognition against Aβ by maintaining canonical Wnt signalling. To test this, we mapped the Dkk1-LRP6 interaction by peptide array and identified a small peptide able to disrupt the Dkk1-LRP6 interaction. This Dkk1-LRP6 'disruptor' peptide dose dependently restores canonical Wnt signalling in the presence of Dkk1; blocks Dkk1-driven dendritic spine loss in primary rat cortical cultures and the accompanying increase in endogenous Aβ production; and when administered intracerebroventricularly to a rat acute Aβ model, blocks Aβ-driven cognitive impairment. These data support our contention that the ability of Aβ to induce Dkk1 and the effects of Dkk1 on LRP6 are an important element in AD aetiopathology and establish Dkk1 as a therapeutic target for protecting synapse and cognition in AD.
Collapse
Affiliation(s)
- Christina Elliott
- King's College London, Centre for Healthy Brain Aging, The Institute of Psychiatry, Psychology and Neuroscience, Denmark Hill, London, UK; Newcastle University, School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle-upon-Tyne, UK
| | - Joshua Jackson
- University of Manchester, Faculty of Biology, Medicine and Health, Division of Pharmacy and Optometry, Manchester, UK
| | - Jane Findlay
- University of Glasgow, School of Cardiovascular and Metabolic Health, Glasgow, Scotland, UK
| | - Gareth Williams
- King's College London, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Anshua Ghosh
- King's College London, Centre for Healthy Brain Aging, The Institute of Psychiatry, Psychology and Neuroscience, Denmark Hill, London, UK
| | - Elena Ribe
- King's College London, Centre for Healthy Brain Aging, The Institute of Psychiatry, Psychology and Neuroscience, Denmark Hill, London, UK
| | | | - Ayesha Khan
- EveBioTek Ltd., Trimble House, Warrington, UK
| | - Clive Ballard
- The University of Exeter Medical School, College of Medicine and Health, St Luke's Campus, Magdalen Road, Exeter EX1 2LU, UK
| | - Dag Aarsland
- King's College London, Centre for Healthy Brain Aging, The Institute of Psychiatry, Psychology and Neuroscience, Denmark Hill, London, UK
| | - George S Baillie
- University of Glasgow, School of Cardiovascular and Metabolic Health, Glasgow, Scotland, UK
| | - Michael Harte
- University of Manchester, Faculty of Biology, Medicine and Health, Division of Pharmacy and Optometry, Manchester, UK
| | - Richard Killick
- King's College London, Centre for Healthy Brain Aging, The Institute of Psychiatry, Psychology and Neuroscience, Denmark Hill, London, UK.
| |
Collapse
|
2
|
Maurice MM, Angers S. Mechanistic insights into Wnt-β-catenin pathway activation and signal transduction. Nat Rev Mol Cell Biol 2025; 26:371-388. [PMID: 39856369 DOI: 10.1038/s41580-024-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/27/2025]
Abstract
In multicellular organisms, Wnt proteins govern stem and progenitor cell renewal and differentiation to regulate embryonic development, adult tissue homeostasis and tissue regeneration. Defects in canonical Wnt signalling, which is transduced intracellularly by β-catenin, have been associated with developmental disorders, degenerative diseases and cancers. Although a simple model describing Wnt-β-catenin signalling is widely used to introduce this pathway and has largely remained unchanged over the past 30 years, in this Review we discuss recent studies that have provided important new insights into the mechanisms of Wnt production, receptor activation and intracellular signalling that advance our understanding of the molecular mechanisms that underlie this important cell-cell communication system. In addition, we review the recent development of molecules capable of activating the Wnt-β-catenin pathway with selectivity in vitro and in vivo that is enabling new lines of study to pave the way for the development of Wnt therapies for the treatment of human diseases.
Collapse
Affiliation(s)
- Madelon M Maurice
- Center for Molecular Medicine, University Medical Center, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| | - Stephane Angers
- Donnelly Centre for Cellular and Biomolecular Research and Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Martinez-Marin D, Stroman GC, Fulton CJ, Pruitt K. Frizzled receptors: gatekeepers of Wnt signaling in development and disease. Front Cell Dev Biol 2025; 13:1599355. [PMID: 40376615 PMCID: PMC12078226 DOI: 10.3389/fcell.2025.1599355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Frizzled (FZD) receptors are a subset of G-protein-coupled receptors (GPCRs), the largest class of human cell surface receptors and a major target of FDA-approved drugs. Activated by Wnt ligands, FZDs regulate key cellular processes such as proliferation, differentiation, and polarity, positioning them at the intersection of developmental biology and disease, including cancer. Despite their significance, FZD signaling remains incompletely understood, particularly in distinguishing receptor-specific roles across canonical and non-canonical Wnt pathways. Challenges include defining ligand-receptor specificity, elucidating signal transduction mechanisms, and understanding the influence of post translational modifications and the cellular context. Structural dynamics, receptor trafficking, and non-canonical signaling contributions also remain areas of active investigation. Recent advances in structural biology, transcriptomics, and functional genomics are beginning to address these gaps, while emerging therapeutic approaches-such as small-molecule modulators and antibodies-highlight the potential of FZDs as drug targets. This review synthesizes current insights into FZD receptor biology, examines ongoing controversies, and outlines promising directions for future research and therapeutic development.
Collapse
Affiliation(s)
| | | | | | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
4
|
Varella FJ, Xavier FAC, Zanirati G, Gonçalves JIB, Previato TTR, Pazzin DB, Pereira-Neto NG, Paglioli E, Martins WA, Palmini A, Dos Santos AS, Abreu JG, Leichtweis KS, Machado DC, Da Costa JC, Marinowic DR. Increased activation of the WNT pathway in brain tissue from patients with cortical dysplasia type IIb. Sci Rep 2025; 15:8049. [PMID: 40055364 PMCID: PMC11889187 DOI: 10.1038/s41598-025-90045-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 05/13/2025] Open
Abstract
Focal cortical dysplasia (FCD) is a malformation of cortical development characterized by a heterogeneous group of lesions with high epileptogenic activity. Somatic mutations in the mTOR pathway are the primary cause of cortical malformations (MCDs). Activation of the WNT pathway inhibits GSK3, which is a key inhibitor of mTOR; consequently, WNT activation is associated with increased activation of the mTOR pathway. Residual samples were obtained from the neocortex of five patients diagnosed with FCD type IIb who underwent surgery. For the control group, residual samples from the neocortex of 3 patients with temporal lobe epilepsy associated with hippocampal sclerosis (TLE-HS) were used. The samples were used to evaluate relative gene expression levels, immunohistochemical characteristics, and the quantification of proteins related to the WNT pathway by Western blot. Gene expression analysis showed increased fold-changes in the genes LRP5, LRP6, DKK1, and DVL1. Immunohistochemistry analysis revealed that the FCD brain samples exhibited more staining for LRP6 compared to control brain tissue. All patients with FCD showed stronger staining for β-catenin. The increased gene expression of WNT pathway genes, combined with the intensified anti-LRP6 antibody staining and increased β-catenin staining, along with the reduced rate of β-catenin phosphorylation observed in patients with FCD, suggests a more pronounced activation of the WNT pathway.
Collapse
Affiliation(s)
- Fabio Jean Varella
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Medicine and Health Sciences, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Medicine and Health Sciences, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biomedical Gerontology, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Medicine and Health Sciences, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Normando G Pereira-Neto
- Epilepsy Surgery Program, São Lucas Hospital, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Eliseu Paglioli
- Epilepsy Surgery Program, São Lucas Hospital, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - William Alves Martins
- Epilepsy Surgery Program, São Lucas Hospital, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Andre Palmini
- Epilepsy Surgery Program, São Lucas Hospital, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - José Garcia Abreu
- Biomedical Science Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kamila Souto Leichtweis
- Biomedical Science Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise C Machado
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaderson Costa Da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
- Graduate Program in Medicine and Health Sciences, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
- Graduate Program in Medicine, Pediatrics and Child Health, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
- Graduate Program in Biomedical Gerontology, Medical School, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
5
|
Abhishek Shah A, Chand D, Ahamad S, Porwal K, Chourasia MK, Mohanan K, Srivastava KR, Chattopadhyay N. Therapeutic targeting of Wnt antagonists by small molecules for treatment of osteoporosis. Biochem Pharmacol 2024; 230:116587. [PMID: 39447984 DOI: 10.1016/j.bcp.2024.116587] [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: 06/05/2024] [Revised: 09/11/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Wnt signaling is one of the key regulators of bone development and homeostasis. Wnt signaling regulates key biological events, including stem cell fate and osteoblast and osteoclast activity, leading to the maintenance of bone mass and strength. Wnt ligands are secreted glycoproteins that bind to Frizzled (FZD) receptors and their coreceptors, lipoprotein receptor-related proteins-5/6 (LRP5/6). Binding of Wnts to FZD triggers canonical (β-catenin-dependent) and noncanonical (β-catenin-independent) pathways. In canonical Wnt signaling, stabilized β-catenin translocates to the nucleus, where it promotes osteoblast differentiation by activating target genes, including Runx2 and Osterix. The negative regulators of Wnt or so-called Wnt antagonists, including CXXC5, sFRP, sclerostin, DKK1, and Notum, compete for Fzd binding, attenuating Wnt signaling. The critical roles of Wnt signaling in bone homeostasis have been established by various bone diseases caused by mutations in Wnt signaling pathways. Loss-of-function mutations in the LRP5 gene cause osteoporosis-pseudoglioma syndrome, whereas gain-of-function mutations are linked to osteopetrosis characterized by high bone density. Sclerosteosis and Van Buchem disease are caused by mutations affecting the SOST gene, which encodes sclerostin, a natural inhibitor of Wnt signalling. Loss-of-function mutations in SOST result in excessive bone growth, markedly increased bone density, and other skeletal abnormalities due to uncontrolled Wnt activity. Considering the clinical relevance of Wnt signaling, targeting Wnt inhibitors is being intensely pursued using small molecules that act by inhibiting endogenous Wnt agonists. We used a computational biology approach to review current data on pharmacophores of Wnt antagonists, assessing their potential as therapeutic candidates for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Aarti Abhishek Shah
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Diwan Chand
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shakir Ahamad
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Konica Porwal
- Division of Endocrinology and Center for Research on Anabolic Skeletal Targets for Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kishor Mohanan
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kinshuk R Srivastava
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research on Anabolic Skeletal Targets for Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
6
|
Ihedioha OC, Marcarian HQ, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major surface components and DKK1 signalling via LRP6 promote migration and longevity of neutrophils in the infection site. Front Immunol 2024; 15:1473133. [PMID: 39502693 PMCID: PMC11534728 DOI: 10.3389/fimmu.2024.1473133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Background Host-related factors highly regulate the increased circulation of neutrophils during Leishmania infection. Platelet-derived Dickkopf-1 (DKK1) is established as a high-affinity ligand to LRP6. Recently, we demonstrated that DKK1 upregulates leukocyte-platelet aggregation, infiltration of neutrophils to the draining lymph node and Th2 differentiation during Leishmania infection, suggesting the potential involvement of the DKK1-LRP6 signalling pathway in neutrophil migration in infectious diseases. Results In this study, we further explored the potential role of DKK1-LRP6 signalling in the migration and longevity of activated neutrophils in the infection site using BALB/c mice with PMNs deficient in LRP6 (LRP6NKO) or BALB/c mice deficient in both PMN LRP6 and platelet DKK1 (LRP6NKO DKK1PKO). Relative to the infected wild-type BALB/c mice, reduced neutrophil activation at the infection site of LRP6NKO or LRP6NKO DKK1PKO mice was noted. The neutrophils obtained from either infected LRP6NKO or LRP6NKO DKK1PKO mice additionally showed a high level of apoptosis. Notably, the level of LRP6 expressing neutrophils was elevated in infected BALB/c mice. Relative to infected BALB/c mice, a significant reduction in parasite load was observed in both LRP6NKO and LRP6NKO DKK1PKO infected mice. Notably, DKK1 levels were comparable in the LRP6NKO and BALB/c mice in response to infection, indicating that PMN activation is the major pathway for DKK1 in promoting parasitemia. Parasite-specific components also play a crucial role in modulating neutrophil circulation in Leishmania disease. Thus, we further determine the contribution of Leishmania membrane components in the migration of neutrophils to the infection site using null mutants deficient in LPG synthesis (Δlpg1- ) or lacking all ether phospholipids (plasmalogens, LPG, and GIPLs) synthesis (Δads1- ). Relative to the WT controls, Δads1- parasite-infected mice showed a sustained decrease in neutrophils and neutrophil-platelet aggregates (for at least 14 days PI), while neutrophils returned to normal in Δlpg1- parasite-infected mice after day 3 PI. Conclusion Our results suggest that DKK1 signalling and Leishmania pathogen-associated molecular patterns appear to regulate the migration and sustenance of viable activated neutrophils in the infection site resulting in chronic type 2 cell-mediated inflammation.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Haley Q. Marcarian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Anutr Sivakoses
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MO, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Infectious Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
7
|
Shetty M, Shenoy S, Amuthan A, Devi V, Kumar N, Kiran A, Shenoy G, Chinta DR, Prasada K S, Shetty A, Rao K G M. Kadukkai maathirai (Indian herbal drug) prevents hepatocellular cancer progression by enhancing GSTM1 expression and modulating β catenin transcription: in-silico and in-vivo study. F1000Res 2024; 13:639. [PMID: 39916986 PMCID: PMC11800331 DOI: 10.12688/f1000research.145961.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/09/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is an aggressive malignancy with poor clinical outcomes. Hence cost-effective drugs with fewer side effects as a standard supportive therapy might yield substantial advantages in efficacy and safety. Kadukkai maathirai (KM) is being used as a supplement in hepatocellular carcinoma. We evaluated whether KM has any preventive action on cancer progression in diethyl nitrosamine (DEN) - induced HCC in rats. Methods DEN was injected to produce HCC in rats, which was confirmed after 16 weeks. All the rats were orally administered KM for 4 weeks. Hepatoprotective potential (serum AST, ALT, ALP, Bilirubin) and anticancer efficacy (body weight, nodule count, tumor progression by histopathology, expression of GSTM1 by Liquid chromatography-mass spectrometry (LC-MS), and In-silico analysis of phytoconstituents against β catenin and LRP analysis were evaluated. Results KM prevented cancer progression against DEN-induced HCC by an increase in GSTM1, a phase II detoxifying enzyme. It significantly reversed altered nodule count, relative liver weight, body weight, and histopathological features of HCC. In silico analysis of phytoconstituents of KM showed that they modulate the intracellular transcription process by inhibiting the armadillo repeat region of β catenin. Conclusions Our results elucidate the potential of KM as a supplement in HCC by reducing nodule count, protecting the liver from further damage, GSTM1 expression, and inhibiting armadillo repeat region of β catenin.
Collapse
Affiliation(s)
- Manjunath Shetty
- Centre Of Excellence, Ocular Nanoscience, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Arul Amuthan
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Vaishali, Bihar, 844102, India
| | - Amruth Kiran
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Ganesh Shenoy
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Diya Rajasekhar Chinta
- Department of Pharmacology, Manipal University College Malaysia, Bukit Baru, Melaka, 75150, Malaysia
| | - Shama Prasada K
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Manipal, Karnataka, 576104, India
| | - Akshatha Shetty
- Department of Research and Development, Muniyal Institute of Ayurveda and Medical Sciences, Manipal, Manipal, Karnataka, 576104, India
| | - Mohandas Rao K G
- Division of Anatomy, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
8
|
Jin X, Wang S, Luo L, Yan F, He Q. Targeting the Wnt/β-catenin signal pathway for the treatment of gastrointestinal cancer: Potential for advancement. Biochem Pharmacol 2024; 227:116463. [PMID: 39102994 DOI: 10.1016/j.bcp.2024.116463] [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: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
Collapse
Affiliation(s)
- Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Fangjie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
9
|
Borges KS, Little DW, Magalhães TDA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, Hammer GD. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609423. [PMID: 39229119 PMCID: PMC11370552 DOI: 10.1101/2024.08.23.609423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
Collapse
Affiliation(s)
- Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald W Little
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chris Lapensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aishwarya Seth
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Svetlana Azova
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Mesut Berber
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amy E O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Adina F Turcu
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dipika R Mohan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Rainey
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
10
|
Xue W, Zhu B, Zhao K, Huang Q, Luo H, Shou Y, Huang Z, Guo H. Targeting LRP6: A new strategy for cancer therapy. Pharmacol Res 2024; 204:107200. [PMID: 38710241 DOI: 10.1016/j.phrs.2024.107200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Targeting specific molecular drivers of tumor growth is a key approach in cancer therapy. Among these targets, the low-density lipoprotein receptor-related protein 6 (LRP6), a vital component of the Wnt signaling pathway, has emerged as an intriguing candidate. As a cell-surface receptor and vital co-receptor, LRP6 is frequently overexpressed in various cancer types, implicating its pivotal role in driving tumor progression. The pursuit of LRP6 as a target for cancer treatment has gained substantial traction, offering a promising avenue for therapeutic intervention. Here, this comprehensive review explores recent breakthroughs in our understanding of LRP6's functions and underlying molecular mechanisms, providing a profound discussion of its involvement in cancer pathogenesis and drug resistance. Importantly, we go beyond discussing LRP6's role in cancer by discussing diverse potential therapeutic approaches targeting this enigmatic protein. These approaches encompass a wide spectrum, including pharmacological agents, natural compounds, non-coding RNAs, epigenetic factors, proteins, and peptides that modulate LRP6 expression or disrupt its interactions. In addition, also discussed the challenges associated with developing LRP6 inhibitors and their advantages over Wnt inhibitors, as well as the drugs that have entered phase II clinical trials. By shedding light on these innovative strategies, we aim to underscore LRP6's significance as a valuable and multifaceted target for cancer treatment, igniting enthusiasm for further research and facilitating translation into clinical applications.
Collapse
Affiliation(s)
- Wei Xue
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Bo Zhu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China
| | - Kaili Zhao
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Qiuju Huang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Hua Luo
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region of China
| | - Yiwen Shou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Zhaoquan Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
11
|
Goto S, Tsutsumi A, Lee Y, Hosojima M, Kabasawa H, Komochi K, Nagatoishi S, Takemoto K, Tsumoto K, Nishizawa T, Kikkawa M, Saito A. Cryo-EM structures elucidate the multiligand receptor nature of megalin. Proc Natl Acad Sci U S A 2024; 121:e2318859121. [PMID: 38771880 PMCID: PMC11145282 DOI: 10.1073/pnas.2318859121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/19/2024] [Indexed: 05/23/2024] Open
Abstract
Megalin (low-density lipoprotein receptor-related protein 2) is a giant glycoprotein of about 600 kDa, mediating the endocytosis of more than 60 ligands, including those of proteins, peptides, and drug compounds [S. Goto, M. Hosojima, H. Kabasawa, A. Saito, Int. J. Biochem. Cell Biol. 157, 106393 (2023)]. It is expressed predominantly in renal proximal tubule epithelial cells, as well as in the brain, lungs, eyes, inner ear, thyroid gland, and placenta. Megalin is also known to mediate the endocytosis of toxic compounds, particularly those that cause renal and hearing disorders [Y. Hori et al., J. Am. Soc. Nephrol. 28, 1783-1791 (2017)]. Genetic megalin deficiency causes Donnai-Barrow syndrome/facio-oculo-acoustico-renal syndrome in humans. However, it is not known how megalin interacts with such a wide variety of ligands and plays pathological roles in various organs. In this study, we elucidated the dimeric architecture of megalin, purified from rat kidneys, using cryoelectron microscopy. The maps revealed the densities of endogenous ligands bound to various regions throughout the dimer, elucidating the multiligand receptor nature of megalin. We also determined the structure of megalin in complex with receptor-associated protein, a molecular chaperone for megalin. The results will facilitate further studies on the pathophysiology of megalin-dependent multiligand endocytic pathways in multiple organs and will also be useful for the development of megalin-targeted drugs for renal and hearing disorders, Alzheimer's disease [B. V. Zlokovic et al., Proc. Natl. Acad. Sci. U.S.A. 93, 4229-4234 (1996)], and other illnesses.
Collapse
Affiliation(s)
- Sawako Goto
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| | - Akihisa Tsutsumi
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Yongchan Lee
- Department of the Biological Membrane Dynamics, Graduate School of Medical Life Science, Yokohama City University, Yokohama230-0045, Japan
| | - Michihiro Hosojima
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| | - Hideyuki Kabasawa
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| | - Koichi Komochi
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| | - Satoru Nagatoishi
- Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo113-8656, Japan
| | - Kazuya Takemoto
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo113-8656, Japan
| | - Tomohiro Nishizawa
- Department of the Biological Membrane Dynamics, Graduate School of Medical Life Science, Yokohama City University, Yokohama230-0045, Japan
| | - Masahide Kikkawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Akihiko Saito
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City951-8510, Japan
| |
Collapse
|
12
|
Roca-Ayats N, Maceda I, Bruque CD, Martínez-Gil N, Garcia-Giralt N, Cozar M, Mellibovsky L, Van Hul W, Lao O, Grinberg D, Balcells S. Evolutionary and functional analyses of LRP5 in archaic and extant modern humans. Hum Genomics 2024; 18:53. [PMID: 38802968 PMCID: PMC11131306 DOI: 10.1186/s40246-024-00616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The human lineage has undergone a postcranial skeleton gracilization (i.e. lower bone mass and strength relative to body size) compared to other primates and archaic populations such as the Neanderthals. This gracilization has been traditionally explained by differences in the mechanical load that our ancestors exercised. However, there is growing evidence that gracilization could also be genetically influenced. RESULTS We have analyzed the LRP5 gene, which is known to be associated with high bone mineral density conditions, from an evolutionary and functional point of view. Taking advantage of the published genomes of archaic Homo populations, our results suggest that this gene has a complex evolutionary history both between archaic and living humans and within living human populations. In particular, we identified the presence of different selective pressures in archaics and extant modern humans, as well as evidence of positive selection in the African and South East Asian populations from the 1000 Genomes Project. Furthermore, we observed a very limited evidence of archaic introgression in this gene (only at three haplotypes of East Asian ancestry out of the 1000 Genomes), compatible with a general erasing of the fingerprint of archaic introgression due to functional differences in archaics compared to extant modern humans. In agreement with this hypothesis, we observed private mutations in the archaic genomes that we experimentally validated as putatively increasing bone mineral density. In particular, four of five archaic missense mutations affecting the first β-propeller of LRP5 displayed enhanced Wnt pathway activation, of which two also displayed reduced negative regulation. CONCLUSIONS In summary, these data suggest a genetic component contributing to the understanding of skeletal differences between extant modern humans and archaic Homo populations.
Collapse
Affiliation(s)
- Neus Roca-Ayats
- Departament de Genètica, Microbiologia i Estadística and IBUB, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Iago Maceda
- CNAG, Centre Nacional d'Analisi Genòmic, C/ Baldiri I Reixach 4, 08028, Barcelona, Spain
- Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carlos David Bruque
- Unidad de Conocimiento Traslacional Hospitalaria Patagónica, Hospital de Alta Complejidad El Calafate - S.A.M.I.C., Santa Cruz, Argentina
| | - Núria Martínez-Gil
- Departament de Genètica, Microbiologia i Estadística and IBUB, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Natàlia Garcia-Giralt
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Departament de Genètica, Microbiologia i Estadística, UB, Barcelona, Spain
| | - Mónica Cozar
- Departament de Genètica, Microbiologia i Estadística and IBUB, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Leonardo Mellibovsky
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp, 2650, Antwerp, Belgium
| | - Oscar Lao
- Institute of Evolutionary Biology, CSIC-Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| | - Daniel Grinberg
- Departament de Genètica, Microbiologia i Estadística and IBUB, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Susanna Balcells
- Departament de Genètica, Microbiologia i Estadística and IBUB, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| |
Collapse
|
13
|
Powell GT, Faro A, Zhao Y, Stickney H, Novellasdemunt L, Henriques P, Gestri G, White ER, Ren J, Lu W, Young RM, Hawkins TA, Cavodeassi F, Schwarz Q, Dreosti E, Raible DW, Li VSW, Wright GJ, Jones EY, Wilson SW. Cachd1 interacts with Wnt receptors and regulates neuronal asymmetry in the zebrafish brain. Science 2024; 384:573-579. [PMID: 38696577 PMCID: PMC7615972 DOI: 10.1126/science.ade6970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/27/2024] [Indexed: 05/04/2024]
Abstract
Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.
Collapse
Affiliation(s)
- Gareth T. Powell
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
| | - Ana Faro
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Heather Stickney
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
- Ambry Genetics; Aliso Viejo, CA 92656, USA
| | - Laura Novellasdemunt
- The Francis Crick Institute; London, NW1 1AT, UK
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology; 08028, Barcelona, Spain
| | - Pedro Henriques
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Gaia Gestri
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | | | - Jingshan Ren
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Rodrigo M. Young
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor; Camino La Piramide 5750, 8580745, Santiago, Chile
| | - Thomas A. Hawkins
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Florencia Cavodeassi
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- St. George’s, University of London; London, SW17 0RE, UK
| | - Quenten Schwarz
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
| | - Elena Dreosti
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - David W. Raible
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
| | | | - Gavin J. Wright
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York; York, YO10 5DD, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Stephen W. Wilson
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| |
Collapse
|
14
|
de Almeida Magalhaes T, Liu J, Chan C, Borges KS, Zhang J, Kane AJ, Wierbowski BM, Ge Y, Liu Z, Mannam P, Zeve D, Weiss R, Breault DT, Huang P, Salic A. Extracellular carriers control lipid-dependent secretion, delivery, and activity of WNT morphogens. Dev Cell 2024; 59:244-261.e6. [PMID: 38154460 PMCID: PMC10872876 DOI: 10.1016/j.devcel.2023.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/29/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
WNT morphogens trigger signaling pathways fundamental for embryogenesis, regeneration, and cancer. WNTs are modified with palmitoleate, which is critical for binding Frizzled (FZD) receptors and activating signaling. However, it is unknown how WNTs are released and spread from cells, given their strong lipid-dependent membrane attachment. We demonstrate that secreted FZD-related proteins and WNT inhibitory factor 1 are WNT carriers, potently releasing lipidated WNTs and forming active soluble complexes. WNT release occurs by direct handoff from the membrane protein WNTLESS to the carriers. In turn, carriers donate WNTs to glypicans and FZDs involved in WNT reception and to the NOTUM hydrolase, which antagonizes WNTs by lipid moiety removal. WNT transfer from carriers to FZDs is greatly facilitated by glypicans that serve as essential co-receptors in Wnt signaling. Thus, an extracellular network of carriers dynamically controls secretion, posttranslational regulation, and delivery of WNT morphogens, with important practical implications for regenerative medicine.
Collapse
Affiliation(s)
| | - Jingjing Liu
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Charlene Chan
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Jiuchun Zhang
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew J Kane
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bradley M Wierbowski
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yunhui Ge
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhiwen Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Prabhath Mannam
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Daniel Zeve
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ron Weiss
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Pengxiang Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adrian Salic
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Fazeli E, Child DD, Bucks SA, Stovarsky M, Edwards G, Rose SE, Yu CE, Latimer C, Kitago Y, Bird T, Jayadev S, Andersen OM, Young JE. A familial missense variant in the Alzheimer's disease gene SORL1 impairs its maturation and endosomal sorting. Acta Neuropathol 2024; 147:20. [PMID: 38244079 PMCID: PMC10799806 DOI: 10.1007/s00401-023-02670-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/11/2023] [Accepted: 12/16/2023] [Indexed: 01/22/2024]
Abstract
The SORL1 gene has recently emerged as a strong Alzheimer's Disease (AD) risk gene. Over 500 different variants have been identified in the gene and the contribution of individual variants to AD development and progression is still largely unknown. Here, we describe a family consisting of 2 parents and 5 offspring. Both parents were affected with dementia and one had confirmed AD pathology with an age of onset > 75 years. All offspring were affected with AD with ages at onset ranging from 53 years to 74 years. DNA was available from the parent with confirmed AD and 5 offspring. We identified a coding variant, p.(Arg953Cys), in SORL1 in 5 of 6 individuals affected by AD. Notably, variant carriers had severe AD pathology, and the SORL1 variant segregated with TDP-43 pathology (LATE-NC). We further characterized this variant and show that this Arginine substitution occurs at a critical position in the YWTD-domain of the SORL1 translation product, SORL1. Functional studies further show that the p.R953C variant leads to retention of the SORL1 protein in the endoplasmic reticulum which leads to decreased maturation and shedding of the receptor and prevents its normal endosomal trafficking. Together, our analysis suggests that p.R953C is a pathogenic variant of SORL1 and sheds light on mechanisms of how missense SORL1 variants may lead to AD.
Collapse
Affiliation(s)
- Elnaz Fazeli
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Daniel D Child
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Stephanie A Bucks
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
| | - Miki Stovarsky
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, 98195, USA
| | - Gabrielle Edwards
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
| | - Shannon E Rose
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Chang-En Yu
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, 98195, USA
- Geriatric Research Education and Clinical Center (GRECC), Veterans Administration Health Care System, Seattle, WA, 98108, USA
| | - Caitlin Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Yu Kitago
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Thomas Bird
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, 98195, USA
- Geriatric Research Education and Clinical Center (GRECC), Veterans Administration Health Care System, Seattle, WA, 98108, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA.
| | - Olav M Andersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| | - Jessica E Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
16
|
Fazeli E, Child DD, Bucks SA, Stovarsky M, Edwards G, Rose SE, Yu CE, Latimer C, Kitago Y, Bird T, Jayadev S, Andersen OM, Young JE. A familial missense variant in the Alzheimer's Disease gene SORL1 impairs its maturation and endosomal sorting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.547348. [PMID: 37461597 PMCID: PMC10349966 DOI: 10.1101/2023.07.01.547348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
The SORL1 gene has recently emerged as a strong Alzheimer's Disease (AD) risk gene. Over 500 different variants have been identified in the gene and the contribution of individual variants to AD development and progression is still largely unknown. Here, we describe a family consisting of 2 parents and 5 offspring. Both parents were affected with dementia and one had confirmed AD pathology with an age of onset >75 years. All offspring were affected with AD with ages at onset ranging from 53yrs-74yrs. DNA was available from the parent with confirmed AD and 5 offspring. We identified a coding variant, p.(Arg953Cys), in SORL1 in 5 of 6 individuals affected by AD. Notably, variant carriers had severe AD pathology, and the SORL1 variant segregated with TDP-43 pathology (LATE-NC). We further characterized this variant and show that this Arginine substitution occurs at a critical position in the YWTD-domain of the SORL1 translation product, SORL1. Functional studies further show that the p.R953C variant leads to retention of the SORL1 protein in the endoplasmic reticulum which leads to decreased maturation and shedding of the receptor and prevents its normal endosomal trafficking. Together, our analysis suggests that p.R953C is a pathogenic variant of SORL1 and sheds light on mechanisms of how missense SORL1 variants may lead to AD.
Collapse
Affiliation(s)
- Elnaz Fazeli
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK8000 AarhusC, Denmark
| | - Daniel D. Child
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Washington USA
| | - Stephanie A. Bucks
- Department of Neurology, University of Washington, Seattle Washington USA
| | - Miki Stovarsky
- Department of Medicine, Division of Medical Genetics University of Washington, Seattle Washington USA
| | - Gabrielle Edwards
- Department of Neurology, University of Washington, Seattle Washington USA
| | - Shannon E. Rose
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Washington USA
| | - Chang-En Yu
- Department of Medicine, Division of Medical Genetics University of Washington, Seattle Washington USA
- Geriatric Research Education and Clinical Center (GRECC), Veterans Administration Health Care System
| | - Caitlin Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Washington USA
| | - Yu Kitago
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Thomas Bird
- Department of Neurology, University of Washington, Seattle Washington USA
- Department of Medicine, Division of Medical Genetics University of Washington, Seattle Washington USA
- Geriatric Research Education and Clinical Center (GRECC), Veterans Administration Health Care System
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle Washington USA
| | - Olav M. Andersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK8000 AarhusC, Denmark
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Washington USA
| |
Collapse
|
17
|
Wen B, Hu S, Yin J, Wu J, Guo W. Molecular Evolution and Protein Structure Variation of Dkk Family. Genes (Basel) 2023; 14:1863. [PMID: 37895211 PMCID: PMC10606412 DOI: 10.3390/genes14101863] [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: 08/24/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Dkks have inhibitory effects on the Wnt signaling pathway, which is involved in the development of skin and its appendages and the regulation of hair growth. The nucleotide sequences were compared and analyzed to further investigate the relationship between the structure and function of the Dkk gene family and vertebrate epidermal hair. The analysis of the molecular evolution of the Dkk family revealed that the evolution rate of the genes changed significantly after speciation, with the Aves and Reptilia branches showing accelerated evolution. Additionally, positive selection was observed at specific sites. The tertiary structure of the protein was also predicted. The analysis of the functional divergence of the Dkk family revealed that the functional divergence coefficient of each gene was greater than 0, with most of the functional divergence sites were located in the Cys-2 domain and a few in the Cys-1 domain. This suggests that the amino acid and functional divergence sites may play a role in regulating the binding of the Dkk family to LRP5/6, and thus affect the inhibition of Wnt signaling, leading to different functions of Dkk1, Dkk2, and Dkk4 in the development of skin hair follicles. In addition, the Dkk families of Aves and Reptilia may have undergone adaptive evolution and functional divergence.
Collapse
Affiliation(s)
- Binhong Wen
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Sile Hu
- College of Life Science, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Jun Yin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Jianghong Wu
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Wenrui Guo
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
18
|
Prajapat M, Singh H, Chaudhary G, Sarma P, Kaur G, Prakash Patel A, Medhi B. A Novel Inhibitor of DKK1/LRP6 Interactions Against the Alzheimer Disease: An Insilco Approach. Bioinform Biol Insights 2023; 17:11779322231183762. [PMID: 37424708 PMCID: PMC10328054 DOI: 10.1177/11779322231183762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/27/2023] [Indexed: 07/11/2023] Open
Abstract
The activation of the Wnt signaling pathway is implicated in a neuroprotective mechanism against the Alzheimer disease. When this pathway is blocked, it activates GSK3 beta, leading to tau hyperphosphorylation and the apoptosis of neurons. Dickkopf-related protein 1 (DKK1) is a protein that competes with the Wnt ligand for the low-density lipoprotein receptor-related protein 6 (LRP6) receptor's binding, interrupting the Wnt-induced Fzd-Wnt-LRP6 complex. This counteracts Wnt's neuroprotective effect and contributes to the progression of the Alzheimer disease. The aim of this study was to use in silico approach to develop new agents that can combat the Alzheimer disease by targeting the interaction between DKK1 and LRP6. To achieve this, we conducted a virtual screening (Vsw) of the Asinex-CNS database library (n = 54 513) compounds against a generated grid in LRP6 protein. From this screening, we selected 6 compounds based on their docking score and performed molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations on the selected ligands. Next, we evaluated the Absorption, Distribution, Metabolism, and Excretion (ADME) results of the 6 screened compounds using the Quick prop module of Schrödinger. We then employed several computational techniques, including PCA (Principal Component Analysis), DCCM (Dynamic Cross-Correlation Map), molecular dynamics simulation, and molecular mechanics/Poisson-Boltzmann surface area (MM/PBSA)-based negative binding free energy (BFE) calculation, to further analyze the compounds. Our extensive computational analysis resulted in the identification of 3 potential hits, LAS 29757582, LAS 29984441, and LAS 29757942. These compounds were found to block the interaction of DKK1 with LRP6 (A and B interface) protein, and their potential as therapeutic agents was supported by negative BFE calculation. Therefore, these compounds show potential as possible therapeutic agents for treating the Alzheimer disease through targeting the interaction between DKK1 and LRP6.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bikash Medhi
- Bikash Medhi, Department of Pharmacology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, Research Block B, 4th Floor, Lab no. 4044, Chandigarh 160012, India.
| |
Collapse
|
19
|
Pokharel SM, Mohanty I, Mariasoosai C, Miura TA, Maddison LA, Natesan S, Bose S. Human beta defensin-3 mediated activation of β-catenin during human respiratory syncytial virus infection: interaction of HBD3 with LDL receptor-related protein 5. Front Microbiol 2023; 14:1186510. [PMID: 37426017 PMCID: PMC10324619 DOI: 10.3389/fmicb.2023.1186510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is a non-segmented negative-sense RNA virus belonging to the paramyxovirus family. RSV infects the respiratory tract to cause pneumonia and bronchiolitis in infants, elderly, and immunocompromised patients. Effective clinical therapeutic options and vaccines to combat RSV infection are still lacking. Therefore, to develop effective therapeutic interventions, it is imperative to understand virus-host interactions during RSV infection. Cytoplasmic stabilization of β-catenin protein results in activation of canonical Wingless (Wnt)/β-catenin signaling pathway that culminates in transcriptional activation of various genes regulated by T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors. This pathway is involved in various biological and physiological functions. Our study shows RSV infection of human lung epithelial A549 cells triggering β-catenin protein stabilization and induction of β-catenin mediated transcriptional activity. Functionally, the activated β-catenin pathway promoted a pro-inflammatory response during RSV infection of lung epithelial cells. Studies with β-catenin inhibitors and A549 cells lacking optimal β-catenin activity demonstrated a significant loss of pro-inflammatory chemokine interleukin-8 (IL-8) release from RSV-infected cells. Mechanistically, our studies revealed a role of extracellular human beta defensin-3 (HBD3) in interacting with cell surface Wnt receptor LDL receptor-related protein-5 (LRP5) to activate the non-canonical Wnt independent β-catenin pathway during RSV infection. We showed gene expression and release of HBD3 from RSV-infected cells and silencing of HBD3 expression resulted in reduced stabilization of β-catenin protein during RSV infection. Furthermore, we observed the binding of extracellular HBD3 with cell surface localized LRP5 protein, and our in silico and protein-protein interaction studies have highlighted a direct interaction of HBD3 with LRP5. Thus, our studies have identified the β-catenin pathway as a key regulator of pro-inflammatory response during RSV infection of human lung epithelial cells. This pathway was induced during RSV infection via a non-canonical Wnt-independent mechanism involving paracrine/autocrine action of extracellular HBD3 activating cell surface Wnt receptor complex by directly interacting with the LRP5 receptor.
Collapse
Affiliation(s)
- Swechha M. Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Indira Mohanty
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Charles Mariasoosai
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Tanya A. Miura
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Lisette A. Maddison
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Senthil Natesan
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| |
Collapse
|
20
|
Gao L, Sun Y, Zhang X, Ma D, Xie A, Wang E, Cheng L, Liu S. Wnt3a-Loaded Extracellular Vesicles Promote Alveolar Epithelial Regeneration after Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206606. [PMID: 37072558 PMCID: PMC10288279 DOI: 10.1002/advs.202206606] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Compromised regeneration resulting from the deactivation of Wnt/β-catenin signaling contributes to the progression of chronic obstructive pulmonary disease (COPD) with limited therapeutic options. Extracellular cytokine-induced Wnt-based signaling provides an alternative option for COPD treatment. However, the hydrophobic nature of Wnt proteins limits their purification and use. This study devises a strategy to deliver the membrane-bound wingless-type MMTV integration site family, member 3A (Wnt3a) over a long distance by anchoring it to the surface of extracellular vesicles (EVs). The newly engineered Wnt3aWG EVs are generated by co-expressing Wnt3a with two genes encoding the membrane protein, WLS, and an engineered glypican, GPC6ΔGPI -C1C2. The bioactivity of Wnt3aWG EVs is validated using a TOPFlash assay and a mesoderm differentiation model of human pluripotent stem cells. Wnt3aWG EVs activate Wnt signaling and promote cell growth following human alveolar epithelial cell injury. In an elastase-induced emphysema model, impaired pulmonary function and enlarged airspace are greatly restored by the intravenous delivery of Wnt3aWG EVs. Single-cell RNA sequencing-based analyses further highlight that Wnt3aWG EV-activated regenerative programs are responsible for its beneficial effects. These findings suggest that EV-based Wnt3a delivery represents a novel therapeutic strategy for lung repair and regeneration after injury.
Collapse
Affiliation(s)
- Lei Gao
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Yongping Sun
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Xinye Zhang
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Ding Ma
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - An Xie
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Enyu Wang
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Linzhao Cheng
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Senquan Liu
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| |
Collapse
|
21
|
Zhang X, Yang G, Liu W, Liu Q, Wang Z, Fan K, Qu F, Huang Y. Screening and Identification of ssDNA Aptamers for Low-Density Lipoprotein (LDL) Receptor-Related Protein 6. Molecules 2023; 28:3838. [PMID: 37175248 PMCID: PMC10180154 DOI: 10.3390/molecules28093838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/15/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6), a member of the low-density lipoprotein receptor (LDLR) family, displays a unique structure and ligand-binding function. As a co-receptor of the Wnt/β-catenin signaling pathway, LRP6 is a novel therapeutic target that plays an important role in the regulation of cardiovascular disease, lipid metabolism, tumorigenesis, and some classical signals. By using capillary electrophoresis-systematic evolution of ligands by exponential enrichment (CE-SELEX), with recombinant human LRP-6 as the target, four candidate aptamers with a stem-loop structure were selected from an ssDNA library-AptLRP6-A1, AptLRP6-A2, AptLRP6-A3, and AptLRP6-A4. The equilibrium dissociation constant KD values between these aptamers and the LRP6 protein were in the range of 0.105 to 1.279 μmol/L, as determined by CE-LIF analysis. Their affinities and specificities were further determined by the gold nanoparticle (AuNP) colorimetric method. Among them, AptLRP6-A3 showed the highest affinity with LRP6-overexpressed human breast cancer cells. Therefore, the LRP6 aptamer identified in this study constitutes a promising modality for the rapid diagnosis and treatment of LRP6-related diseases.
Collapse
Affiliation(s)
- Xiaomin Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Ge Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Wenjing Liu
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101125, China
| | - Qing Liu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Qu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
22
|
Li L, Zeng P, Yu L, Yang J, Man J, Zhou L, Zhao L. Salinomycin sodium exerts anti diffuse large B-cell lymphoma activity through inhibition of LRP6-mediated Wnt/β-catenin and mTORC1 signaling. Leuk Lymphoma 2023:1-10. [PMID: 37092573 DOI: 10.1080/10428194.2023.2202291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Low-density lipoprotein receptor-related protein-6 (LRP6) is overexpressed in various cancers. The small molecule salinomycin sodium inhibits LRP6. We observed a higher proportion of subjects with non-germinal center B (non-GCB) subtypes having high LRP6 expression than those with GCB subtypes by immunohistochemistry. The PCR and Western blot assays demonstrated increased LRP6 expression in non-GCB subtype cells. In addition, CCK-8 assays and transwell cell migration assays revealed that salinomycin sodium exhibited dose- and time-dependent inhibition of proliferation and migration in non-GCB subtype cells. Furthermore, Western blot assays showed that salinomycin sodium decreased the expression of Bcl2, while increasing the expression of Bax. Additionally, salinomycin sodium suppressed LRP6 expression, blocked LRP6 phosphorylation, and inhibited the Wnt/β-catenin and mTORC1 signaling pathways. Our results suggest that LRP6 is highly expressed in non-GCB subtype. Furthermore, salinomycin sodium inhibited LRP6 expression and the Wnt/β-catenin and mTORC1 signaling in non-GCB subtype cells, and displayed potent anticancer activity.
Collapse
Affiliation(s)
- Liangliang Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Pengyun Zeng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Lili Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Jincai Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Jiancheng Man
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Lanxia Zhou
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, P.R. China
| | - Li Zhao
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, Gansu, P.R. China
| |
Collapse
|
23
|
Tsutsumi N, Hwang S, Waghray D, Hansen S, Jude KM, Wang N, Miao Y, Glassman CR, Caveney NA, Janda CY, Hannoush RN, Garcia K. Structure of the Wnt-Frizzled-LRP6 initiation complex reveals the basis for coreceptor discrimination. Proc Natl Acad Sci U S A 2023; 120:e2218238120. [PMID: 36893265 PMCID: PMC10089208 DOI: 10.1073/pnas.2218238120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/09/2023] [Indexed: 03/11/2023] Open
Abstract
Wnt morphogens are critical for embryonic development and tissue regeneration. Canonical Wnts form ternary receptor complexes composed of tissue-specific Frizzled (Fzd) receptors together with the shared LRP5/6 coreceptors to initiate β-catenin signaling. The cryo-EM structure of a ternary initiation complex of an affinity-matured XWnt8-Frizzled8-LRP6 complex elucidates the basis of coreceptor discrimination by canonical Wnts by means of their N termini and linker domains that engage the LRP6 E1E2 domain funnels. Chimeric Wnts bearing modular linker "grafts" were able to transfer LRP6 domain specificity between different Wnts and enable non-canonical Wnt5a to signal through the canonical pathway. Synthetic peptides comprising the linker domain serve as Wnt-specific antagonists. The structure of the ternary complex provides a topological blueprint for the orientation and proximity of Frizzled and LRP6 within the Wnt cell surface signalosome.
Collapse
Affiliation(s)
- Naotaka Tsutsumi
- HHMI, Stanford University School of Medicine, Stanford, CA94305
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama700-8530, Japan
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA94080
| | - Deepa Waghray
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Simon Hansen
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA94080
| | - Kevin M. Jude
- HHMI, Stanford University School of Medicine, Stanford, CA94305
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Nan Wang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Yi Miao
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Caleb R. Glassman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Nathanael A. Caveney
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Claudia Y. Janda
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
- Princess Máxima Center for Pediatric Oncology, 3584 CSUtrecht, Netherlands
| | - Rami N. Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA94080
| | - K. Christopher Garcia
- HHMI, Stanford University School of Medicine, Stanford, CA94305
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
24
|
N-Glycosylation of LRP6 by B3GnT2 Promotes Wnt/β-Catenin Signalling. Cells 2023; 12:cells12060863. [PMID: 36980204 PMCID: PMC10047360 DOI: 10.3390/cells12060863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Reception of Wnt signals by cells is predominantly mediated by Frizzled receptors in conjunction with a co-receptor, the latter being LRP6 or LRP5 for the Wnt/β-catenin signalling pathway. It is important that cells maintain precise control of receptor activation events in order to properly regulate Wnt/β-catenin signalling as aberrant signalling can result in disease in humans. Phosphorylation of the intracellular domain (ICD) of LRP6 is well known to regulate Wntβ-catenin signalling; however, less is known for regulatory post-translational modification events within the extracellular domain (ECD). Using a cell culture-based expression screen for functional regulators of LRP6, we identified a glycosyltransferase, B3GnT2-like, from a teleost fish (medaka) cDNA library, that modifies LRP6 and regulates Wnt/β-catenin signalling. We provide both gain-of-function and loss-of-function evidence that the single human homolog, B3GnT2, promotes extension of polylactosamine chains at multiple N-glycans on LRP6, thereby enhancing trafficking of LRP6 to the plasma membrane and promoting Wnt/β-catenin signalling. Our findings further highlight the importance of LRP6 as a regulatory hub in Wnt signalling and provide one of the few examples of how a specific glycosyltransferase appears to selectively target a signalling pathway component to alter cellular signalling events.
Collapse
|
25
|
Thakur AK, Miller SE, Liau NPD, Hwang S, Hansen S, de Sousa E Melo F, Sudhamsu J, Hannoush RN. Synthetic Multivalent Disulfide-Constrained Peptide Agonists Potentiate Wnt1/β-Catenin Signaling via LRP6 Coreceptor Clustering. ACS Chem Biol 2023; 18:772-784. [PMID: 36893429 DOI: 10.1021/acschembio.2c00753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Wnt ligands are critical for tissue homeostasis and form a complex with LRP6 and frizzled coreceptors to initiate Wnt/β-catenin signaling. Yet, how different Wnts achieve various levels of signaling activation through distinct domains on LRP6 remains elusive. Developing tool ligands that target individual LRP6 domains could help elucidate the mechanism of Wnt signaling regulation and uncover pharmacological approaches for pathway modulation. We employed directed evolution of a disulfide constrained peptide (DCP) to identify molecules that bind to the third β-propeller domain of LRP6. The DCPs antagonize Wnt3a while sparing Wnt1 signaling. Using PEG linkers with different geometries, we converted the Wnt3a antagonist DCPs to multivalent molecules that potentiated Wnt1 signaling by clustering the LRP6 coreceptor. The mechanism of potentiation is unique as it occurred only in the presence of extracellular secreted Wnt1 ligand. While all DCPs recognized a similar binding interface on LRP6, they displayed different spatial orientations that influenced their cellular activities. Moreover, structural analyses revealed that the DCPs exhibited new folds that were distinct from the parent DCP framework they were evolved from. The multivalent ligand design principles highlighted in this study provide a path for developing peptide agonists that modulate different branches of cellular Wnt signaling.
Collapse
Affiliation(s)
- Avinash K Thakur
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Stephen E Miller
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Nicholas P D Liau
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Simon Hansen
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Felipe de Sousa E Melo
- Department of Molecular Oncology, Genentech, South San Francisco, California 94080, United States
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
26
|
Ruiz-Gómez G, Salbach-Hirsch J, Dürig JN, Köhler L, Balamurugan K, Rother S, Heidig SL, Moeller S, Schnabelrauch M, Furesi G, Pählig S, Guillem-Gloria PM, Hofbauer C, Hintze V, Pisabarro MT, Rademann J, Hofbauer LC. Rational engineering of glycosaminoglycan-based Dickkopf-1 scavengers to improve bone regeneration. Biomaterials 2023; 297:122105. [PMID: 37031548 DOI: 10.1016/j.biomaterials.2023.122105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The WNT signaling pathway is a central regulator of bone development and regeneration. Functional alterations of WNT ligands and inhibitors are associated with a variety of bone diseases that affect bone fragility and result in a high medical and socioeconomic burden. Hence, this cellular pathway has emerged as a novel target for bone-protective therapies, e.g. in osteoporosis. Here, we investigated glycosaminoglycan (GAG) recognition by Dickkopf-1 (DKK1), a potent endogenous WNT inhibitor, and the underlying functional implications in order to develop WNT signaling regulators. In a multidisciplinary approach we applied in silico structure-based de novo design strategies and molecular dynamics simulations combined with synthetic chemistry and surface plasmon resonance spectroscopy to Rationally Engineer oligomeric Glycosaminoglycan derivatives (REGAG) with improved neutralizing properties for DKK1. In vitro and in vivo assays show that the GAG modification to obtain REGAG translated into increased WNT pathway activity and improved bone regeneration in a mouse calvaria defect model with critical size bone lesions. Importantly, the developed REGAG outperformed polymeric high-sulfated hyaluronan (sHA3) in enhancing bone healing up to 50% due to their improved DKK1 binding properties. Thus, rationally engineered GAG variants may represent an innovative strategy to develop novel therapeutic approaches for regenerative medicine.
Collapse
Affiliation(s)
- Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47/51, D-01307, Dresden, Germany
| | - Juliane Salbach-Hirsch
- Division of Endocrinology, Diabetes and Bone Diseases & Center for Healthy Aging, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstraße 74, D-01307, Dresden, Germany
| | - Jan-Niklas Dürig
- Institute of Pharmacy - Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195, Berlin, Germany
| | - Linda Köhler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069, Dresden, Germany
| | - Kanagasabai Balamurugan
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47/51, D-01307, Dresden, Germany
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069, Dresden, Germany
| | - Sophie-Luise Heidig
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47/51, D-01307, Dresden, Germany
| | | | | | - Giulia Furesi
- Division of Endocrinology, Diabetes and Bone Diseases & Center for Healthy Aging, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstraße 74, D-01307, Dresden, Germany
| | - Sophie Pählig
- Division of Endocrinology, Diabetes and Bone Diseases & Center for Healthy Aging, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstraße 74, D-01307, Dresden, Germany
| | - Pedro M Guillem-Gloria
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47/51, D-01307, Dresden, Germany
| | - Christine Hofbauer
- National Center for Tumor Diseases/University Cancer Center Dresden, Technische Universität Dresden Medical Center, Fetscherstraße 74, D-01307, Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069, Dresden, Germany.
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47/51, D-01307, Dresden, Germany.
| | - Jörg Rademann
- Institute of Pharmacy - Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195, Berlin, Germany.
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes and Bone Diseases & Center for Healthy Aging, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstraße 74, D-01307, Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, D-01307, Dresden, Germany.
| |
Collapse
|
27
|
Gradauskaite V, Inglebert M, Doench J, Scherer M, Dettwiler M, Wyss M, Shrestha N, Rottenberg S, Plattet P. LRP6 Is a Functional Receptor for Attenuated Canine Distemper Virus. mBio 2023; 14:e0311422. [PMID: 36645301 PMCID: PMC9973313 DOI: 10.1128/mbio.03114-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/06/2022] [Indexed: 01/17/2023] Open
Abstract
Wild-type canine distemper virus (CDV) is an important pathogen of dogs as well as wildlife that can infect immune and epithelial cells through two known receptors: the signaling lymphocytic activation molecule (SLAM) and nectin-4, respectively. Conversely, the ferret and egg-adapted CDV-Onderstepoort strain (CDV-OP) is employed as an effective vaccine for dogs. CDV-OP also exhibits promising oncolytic properties, such as its abilities to infect and kill multiple cancer cells in vitro. Interestingly, several cancer cells do not express SLAM or nectin-4, suggesting the presence of a yet unknown entry factor for CDV-OP. By conducting a genome-wide CRISPR/Cas9 knockout (KO) screen in CDV-OP-susceptible canine mammary carcinoma P114 cells, which neither express SLAM nor nectin-4, we identified low-density lipoprotein receptor-related protein 6 (LRP6) as a host factor that promotes CDV-OP infectivity. Whereas the genetic ablation of LRP6 rendered cells resistant to infection, ectopic expression in resistant LRP6KO cells restored susceptibility. Furthermore, multiple functional studies revealed that (i) the overexpression of LRP6 leads to increased cell-cell fusion, (ii) a soluble construct of the viral receptor-binding protein (solHOP) interacts with a soluble form of LRP6 (solLRP6), (iii) an H-OP point mutant that prevents interaction with solLRP6 abrogates cell entry in multiple cell lines once transferred into recombinant viral particles, and (iv) vesicular stomatitis virus (VSV) pseudotyped with CDV-OP envelope glycoproteins loses its infectivity in LRP6KO cells. Collectively, our study identified LRP6 as the long sought-after cell entry receptor of CDV-OP in multiple cell lines, which set the molecular bases to refine our understanding of viral-cell adaptation and to further investigate its oncolytic properties. IMPORTANCE Oncolytic viruses (OV) have gathered increasing interest in recent years as an alternative option to treat cancers. The Onderstepoort strain of canine distemper virus (CDV-OP), an enveloped RNA virus belonging to the genus Morbillivirus, is employed as a safe and efficient vaccine for dogs against distemper disease. Importantly, although CDV-OP can infect and kill multiple cancer cell lines, the basic mechanisms of entry remain to be elucidated, as most of those transformed cells do not express natural receptors (i.e., SLAM and nectin-4). In this study, using a genome-wide CRISPR/Cas9 knockout screen, we describe the discovery of LRP6 as a novel functional entry receptor for CDV-OP in various cancer cell lines and thereby uncover a basic mechanism of cell culture adaptation. Since LRP6 is upregulated in various cancer types, our data provide important insights in order to further investigate the oncolytic properties of CDV-OP.
Collapse
Affiliation(s)
- Vaiva Gradauskaite
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Marine Inglebert
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - John Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Melanie Scherer
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Martina Dettwiler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marianne Wyss
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Neeta Shrestha
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
| | - Philippe Plattet
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Beenken A, Cerutti G, Brasch J, Guo Y, Sheng Z, Erdjument-Bromage H, Aziz Z, Robbins-Juarez SY, Chavez EY, Ahlsen G, Katsamba PS, Neubert TA, Fitzpatrick AWP, Barasch J, Shapiro L. Structures of LRP2 reveal a molecular machine for endocytosis. Cell 2023; 186:821-836.e13. [PMID: 36750096 PMCID: PMC9993842 DOI: 10.1016/j.cell.2023.01.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023]
Abstract
The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2 or megalin) is representative of the phylogenetically conserved subfamily of giant LDL receptor-related proteins, which function in endocytosis and are implicated in diseases of the kidney and brain. Here, we report high-resolution cryoelectron microscopy structures of LRP2 isolated from mouse kidney, at extracellular and endosomal pH. The structures reveal LRP2 to be a molecular machine that adopts a conformation for ligand binding at the cell surface and for ligand shedding in the endosome. LRP2 forms a homodimer, the conformational transformation of which is governed by pH-sensitive sites at both homodimer and intra-protomer interfaces. A subset of LRP2 deleterious missense variants in humans appears to impair homodimer assembly. These observations lay the foundation for further understanding the function and mechanism of LDL receptors and implicate homodimerization as a conserved feature of the LRP receptor subfamily.
Collapse
Affiliation(s)
- Andrew Beenken
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Gabriele Cerutti
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Julia Brasch
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Zainab Aziz
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Estefania Y Chavez
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Goran Ahlsen
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Phinikoula S Katsamba
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony W P Fitzpatrick
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Columbia University George M. O'Brien Urology Center, New York, NY 10032, USA.
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
29
|
Xu M, Chen X, Yu Z, Li X. Receptors that bind to PEDF and their therapeutic roles in retinal diseases. Front Endocrinol (Lausanne) 2023; 14:1116136. [PMID: 37139333 PMCID: PMC10149954 DOI: 10.3389/fendo.2023.1116136] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
Retinal neovascular, neurodegenerative, and inflammatory diseases represented by diabetic retinopathy are the main types of blinding eye disorders that continually cause the increased burden worldwide. Pigment epithelium-derived factor (PEDF) is an endogenous factor with multiple effects including neurotrophic activity, anti-angiogenesis, anti-tumorigenesis, and anti-inflammatory activity. PEDF activity depends on the interaction with the proteins on the cell surface. At present, seven independent receptors, including adipose triglyceride lipase, laminin receptor, lipoprotein receptor-related protein, plexin domain-containing 1, plexin domain-containing 2, F1-ATP synthase, and vascular endothelial growth factor receptor 2, have been demonstrated and confirmed to be high affinity receptors for PEDF. Understanding the interactions between PEDF and PEDF receptors, their roles in normal cellular metabolism and the response the initiate in disease will be accommodating for elucidating the ways in which inflammation, angiogenesis, and neurodegeneration exacerbate disease pathology. In this review, we firstly introduce PEDF receptors comprehensively, focusing particularly on their expression pattern, ligands, related diseases, and signal transduction pathways, respectively. We also discuss the interactive ways of PEDF and receptors to expand the prospective understanding of PEDF receptors in the diagnosis and treatment of retinal diseases.
Collapse
|
30
|
Timberlake AT, Kiziltug E, Jin SC, Nelson-Williams C, Loring E, Allocco A, Marlier A, Banka S, Stuart H, Passos-Buenos MR, Rosa R, Rogatto SR, Tonne E, Stiegler AL, Boggon TJ, Alperovich M, Steinbacher D, Staffenberg DA, Flores RL, Persing JA, Kahle KT, Lifton RP. De novo mutations in the BMP signaling pathway in lambdoid craniosynostosis. Hum Genet 2023; 142:21-32. [PMID: 35997807 DOI: 10.1007/s00439-022-02477-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 01/18/2023]
Abstract
Lambdoid craniosynostosis (CS) is a congenital anomaly resulting from premature fusion of the cranial suture between the parietal and occipital bones. Predominantly sporadic, it is the rarest form of CS and its genetic etiology is largely unexplored. Exome sequencing of 25 kindreds, including 18 parent-offspring trios with sporadic lambdoid CS, revealed a marked excess of damaging (predominantly missense) de novo mutations that account for ~ 40% of sporadic cases. These mutations clustered in the BMP signaling cascade (P = 1.6 × 10-7), including mutations in genes encoding BMP receptors (ACVRL1 and ACVR2A), transcription factors (SOX11, FOXO1) and a transcriptional co-repressor (IFRD1), none of which have been implicated in other forms of CS. These missense mutations are at residues critical for substrate or target sequence recognition and many are inferred to cause genetic gain-of-function. Additionally, mutations in transcription factor NFIX were implicated in syndromic craniosynostosis affecting diverse sutures. Single cell RNA sequencing analysis of the mouse lambdoid suture identified enrichment of mutations in osteoblast precursors (P = 1.6 × 10-6), implicating perturbations in the balance between proliferation and differentiation of osteoprogenitor cells in lambdoid CS. The results contribute to the growing knowledge of the genetics of CS, have implications for genetic counseling, and further elucidate the molecular etiology of premature suture fusion.
Collapse
Affiliation(s)
- Andrew T Timberlake
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA.
| | - Emre Kiziltug
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Sheng Chih Jin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | | | - Erin Loring
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | | | - August Allocco
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Siddharth Banka
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9WL, UK.,Manchester Centre for Genomic Medicine, Health Innovation Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Helen Stuart
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9WL, UK.,Manchester Centre for Genomic Medicine, Health Innovation Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | | | - Rafael Rosa
- Clinical Genetics, UFCSPA and Irmandade da Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, RS, Brazil
| | - Silvia R Rogatto
- Neogene Laboratory, Research Center (CIPE), AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Elin Tonne
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.,University of Oslo, Oslo, Norway
| | - Amy L Stiegler
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Titus J Boggon
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Michael Alperovich
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Derek Steinbacher
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - David A Staffenberg
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - Roberto L Flores
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - John A Persing
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, MA, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA. .,Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
31
|
Giatagana EM, Berdiaki A, Gaardløs M, Tsatsakis AM, Samsonov SA, Nikitovic D. Rapamycin-induced autophagy in osteosarcoma cells is mediated via the biglycan/Wnt/β-catenin signaling axis. Am J Physiol Cell Physiol 2022; 323:C1740-C1756. [PMID: 36280393 DOI: 10.1152/ajpcell.00368.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biglycan is a class I secreted small leucine-rich proteoglycan (SLRP), which regulates signaling pathways connected to bone pathologies. Autophagy is a vital catabolic process with a dual role in cancer progression. Here, we show that biglycan inhibits autophagy in two osteosarcoma cell lines (P ≤ 0.001), while rapamycin-induced autophagy decreases biglycan expression in MG63 osteosarcoma cells and abrogates the biglycan-induced cell growth increase (P ≤ 0.001). Rapamycin also inhibits β-catenin translocation to the nucleus, inhibiting the Wnt pathway (P ≤ 0.001) and reducing biglycan's colocalization with the Wnt coreceptor LRP6 (P ≤ 0.05). Furthermore, biglycan exhibits protective effects against the chemotherapeutic drug doxorubicin in MG63 OS cells through an autophagy-dependent manner (P ≤ 0.05). Cotreatment of these cells with rapamycin and doxorubicin enhances cells response to doxorubicin by decreasing biglycan (P ≤ 0.001) and β-catenin (P ≤ 0.05) expression. Biglycan deficiency leads to increased caspase-3 activation (P ≤ 0.05), suggesting increased apoptosis of biglycan-deficient cells treated with doxorubicin. Computational models of LRP6 and biglycan complexes suggest that biglycan changes the receptor's ability to interact with other signaling molecules by affecting the interdomain bending angles in the receptor structure. Biglycan binding to LRP6 activates the Wnt pathway and β-catenin nuclear translocation by disrupting β-catenin degradation complex (P ≤ 0.01 and P ≤ 0.05). Interestingly, this mechanism is not followed in moderately differentiated, biglycan-nonexpressing U-2OS OS cells. To sum up, biglycan exhibits protective effects against the doxorubicin in MG63 OS cells by activating the Wnt signaling pathway and inhibiting autophagy.
Collapse
Affiliation(s)
- Eirini-Maria Giatagana
- Laboratory of Histology-Embryology, Medical School, University of Crete, Heraklion Greece
| | - Aikaterini Berdiaki
- Laboratory of Histology-Embryology, Medical School, University of Crete, Heraklion Greece
| | - Margrethe Gaardløs
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Aristidis M Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, Heraklion, Greece
| | - Sergey A Samsonov
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, Medical School, University of Crete, Heraklion Greece
| |
Collapse
|
32
|
Manandhar S, Sankhe R, Priya K, Hari G, Kumar B H, Mehta CH, Nayak UY, Pai KSR. Molecular dynamics and structure-based virtual screening and identification of natural compounds as Wnt signaling modulators: possible therapeutics for Alzheimer's disease. Mol Divers 2022; 26:2793-2811. [PMID: 35146638 PMCID: PMC9532339 DOI: 10.1007/s11030-022-10395-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/22/2022] [Indexed: 11/29/2022]
Abstract
Wnt signaling pathway is an evolutionarily conserved pathway responsible for neurogenesis, axon outgrowth, neuronal polarity, synapse formation, and maintenance. Downregulation of Wnt signaling has been found in patients with Alzheimer's disease (AD). Several experimental approaches to activate Wnt signaling pathway have proven to be beneficial in alleviating AD, which is one of the new therapeutic approaches for AD. The current study focuses on the computational structure-based virtual screening followed by the identification of potential phytomolecules targeting different markers of Wnt signaling like WIF1, DKK1, LRP6, GSK-3β, and acetylcholine esterase. Initially, screening of 1924 compounds from the plant-based library of Zinc database was done for the selected five proteins using docking approach followed by MM-GBSA calculations. The top five hit molecules were identified for each protein. Based on docking score, and binding interactions, the top two hit molecules for each protein were selected as promising molecules for the molecular dynamic (MD) simulation study with the five proteins. Therefore, from this in silico based study, we report that Mangiferin could be a potential molecule targeting Wnt signaling pathway modulating the LRP6 activity, Baicalin for AChE activity, Chebulic acid for DKK1, ZINC103539689 for WIF1, and Morin for GSk-3β protein. However, further validation of the activity is warranted based on in vivo and in vitro experiments for better understanding and strong claim. This study provides an in silico approach for the identification of modulators of the Wnt signaling pathway as a new therapeutic approach for AD.
Collapse
Affiliation(s)
- Suman Manandhar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Keerthi Priya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Gangadhar Hari
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Harish Kumar B
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
33
|
Mahoney JP, Bruguera ES, Vasishtha M, Killingsworth LB, Kyaw S, Weis WI. PI(4,5)P 2-stimulated positive feedback drives the recruitment of Dishevelled to Frizzled in Wnt-β-catenin signaling. Sci Signal 2022; 15:eabo2820. [PMID: 35998232 PMCID: PMC9528458 DOI: 10.1126/scisignal.abo2820] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the Wnt-β-catenin pathway, Wnt binding to Frizzled (Fzd) and LRP5 or LRP6 (LRP5/6) co-receptors inhibits the degradation of the transcriptional coactivator β-catenin by recruiting the cytosolic effector Dishevelled (Dvl). Polymerization of Dvl at the plasma membrane recruits the β-catenin destruction complex, enabling the phosphorylation of LRP5/6, a key step in inhibiting β-catenin degradation. Using purified Fzd proteins reconstituted in lipid nanodiscs, we investigated the factors that promote the recruitment of Dvl to the plasma membrane. We found that the affinity of Fzd for Dvl was not affected by Wnt ligands, in contrast to other members of the GPCR superfamily for which the binding of extracellular ligands affects the affinity for downstream transducers. Instead, Fzd-Dvl binding was enhanced by increased concentration of the lipid PI(4,5)P2, which is generated by Dvl-associated lipid kinases in response to Wnt and which is required for LRP5/6 phosphorylation. Moreover, binding to Fzd did not promote Dvl DEP domain dimerization, which has been proposed to be required for signaling downstream of Fzd. Our findings suggest a positive feedback loop in which Wnt-stimulated local PI(4,5)P2 production enhances Dvl recruitment and further PI(4,5)P2 production to support Dvl polymerization, LRP5/6 phosphorylation, and β-catenin stabilization.
Collapse
Affiliation(s)
- Jacob P Mahoney
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Elise S Bruguera
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Mansi Vasishtha
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Lauren B Killingsworth
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Saw Kyaw
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - William I Weis
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| |
Collapse
|
34
|
Kantaputra P, Jatooratthawichot P, Jintakanon K, Intachai W, Pradermdutsadeeporn P, Adisornkanj P, Tongsima S, Ngamphiw C, Olsen B, Tucker AS, Cairns JRK. Mutations in LRP6 highlight the role of WNT signalling in oral exostoses and dental anomalies. Arch Oral Biol 2022; 142:105514. [DOI: 10.1016/j.archoralbio.2022.105514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 07/28/2022] [Indexed: 02/07/2023]
|
35
|
Jiang H, Zhang Z, Yu Y, Chu HY, Yu S, Yao S, Zhang G, Zhang BT. Drug Discovery of DKK1 Inhibitors. Front Pharmacol 2022; 13:847387. [PMID: 35355709 PMCID: PMC8959454 DOI: 10.3389/fphar.2022.847387] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Dickkopf-1 (DKK1) is a well-characterized Wnt inhibitor and component of the Wnt/β-catenin signaling pathway, whose dysregulation is associated with multiple abnormal pathologies including osteoporosis, Alzheimer's disease, diabetes, and various cancers. The Wnt signaling pathway has fundamental roles in cell fate determination, cell proliferation, and survival; thus, its mis-regulation can lead to disease. Although DKK1 is involved in other signaling pathways, including the β-catenin-independent Wnt pathway and the DKK1/CKAP4 pathway, the inhibition of DKK1 to propagate Wnt/β-catenin signals has been validated as an effective way to treat related diseases. In fact, strategies for developing DKK1 inhibitors have produced encouraging clinical results in different pathological models, and many publications provide detailed information about these inhibitors, which include small molecules, antibodies, and nucleic acids, and may function at the protein or mRNA level. However, no systematic review has yet provided an overview of the various aspects of their development and prospects. Therefore, we review the DKK1 inhibitors currently available or under study and provide an outlook on future studies involving DKK1 and drug discovery.
Collapse
Affiliation(s)
- Hewen Jiang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Zongkang Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Yuanyuan Yu
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hang Yin Chu
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Sifan Yu
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Shanshan Yao
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Ge Zhang
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| |
Collapse
|
36
|
LRP6 Receptor Plays Essential Functions in Development and Human Diseases. Genes (Basel) 2022; 13:genes13010120. [PMID: 35052459 PMCID: PMC8775365 DOI: 10.3390/genes13010120] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
LRP6 is a member of the low-density lipoprotein receptor superfamily of cell-surface receptors. It is required for the activation of the Wnt/β-catenin signalling pathway. LRP6 is detected in different tissue types and is involved in numerous biological activities such as cell proliferation, specification, metastatic cancer, and embryonic development. LRP6 is essential for the proper development of different organs in vertebrates, such as Xenopus laevis, chickens, and mice. In human, LRP6 overexpression and mutations have been reported in multiple complex diseases including hypertension, atherosclerosis, and cancers. Clinical studies have shown that LRP6 is involved in various kinds of cancer, such as bladder and breast cancer. Therefore, in this review, we focus on the structure of LRP6 and its interactions with Wnt inhibitors (DKK1, SOST). We also discuss the expression of LRP6 in different model systems, with emphasis on its function in development and human diseases.
Collapse
|
37
|
Azadian Z, Hosseini S, Dizjikan ZP, Kazemi J, Marzouni ET, Wang PY, Alipour A, Shahsavarani H. Computational and in vitro validation of cardiogenic induction of quercetin on adipose-derived mesenchymal stromal cells through the inhibition of Wnt and non-Smad-dependent TGF-β pathways. J Cell Biochem 2021; 123:450-468. [PMID: 34825407 DOI: 10.1002/jcb.30189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/25/2021] [Accepted: 11/13/2021] [Indexed: 12/11/2022]
Abstract
Exploiting human mesenchymal stem cells (hMSCs) was proposed as a promising therapeutic approach for cardiovascular disease due to their capacity to differentiate into cardiac cells. Though modulation of the intracellular signaling pathways dominantly WNT/β catenin and transforming growth factor-β (TGF-β) have been reported to promote differentiation of hMSCs into cardiomyocytes in the prevailing literature, a safe and reproducible system for their clinical application has not yet turned into reality. In the present study, the molecular docking-based strategy was first applied for evaluating the potency of some natural phenolic compounds in the modulation of Wnt and TGF-β signaling pathways using a vital class of crystallographic protein structures of WNT signaling regulators such as Frizzled, Disheveled, GSK3-β, β-catenin, LRP 5/6 extracellular domain, Tankyrase and their variety of active pockets. Then, the impacts of plant-derived chemical compounds on the regulation of the relevant signals for the differentiation of hMSCs into the definitive mesoderm lineage and cardiac progenitors were assessed in vitro. Data obtained revealed the synergistic activity of Wnt and TGF-β superfamily to direct cardiac differentiation in human cardiogenesis by comparing cardiac gene expression in the presence and absence of the TGF-β inhibitors. We found that the inhibitory effect of canonical Wnt/β-catenin is sufficient to cause proper cardiomyocyte differentiation, but the TGF-β pathway plays a vital role in enhancing the expression of the cardiomyocyte-specific marker (cTnT). It was found that quercetin, a p38MAPK inhibitor with the high energy dock to the active pocket of Wnt receptors, promotes cardiac differentiation via the inhibition of both Wnt and non-Smad TGF-β pathways. Altogether, data presented here can contribute to the development of a feasible and efficient cardiac differentiation protocol as an "off-the-shelf" therapeutic source using novel natural agents for cardiac repair or regeneration.
Collapse
Affiliation(s)
- Zahra Azadian
- Department of Cell and Molecular Sciences, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.,Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Saadi Hosseini
- Department of Cell and Molecular Sciences, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.,Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Zohre Panahi Dizjikan
- Department of Cell and Molecular Sciences, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Javad Kazemi
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Eisa Tahmasbpour Marzouni
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Peng-Yuan Wang
- Stem Cell Bioengineering Lab, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur institute of Iran, Tehran, Iran
| | - Hosein Shahsavarani
- Department of Cell and Molecular Sciences, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.,Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
38
|
Chu KY, Wang YL, Chou YR, Chen JT, Wang YP, Simmer JP, Hu JCC, Wang SK. Synergistic Mutations of LRP6 and WNT10A in Familial Tooth Agenesis. J Pers Med 2021; 11:jpm11111217. [PMID: 34834569 PMCID: PMC8621929 DOI: 10.3390/jpm11111217] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 02/07/2023] Open
Abstract
Familial tooth agenesis (FTA), distinguished by developmental failure of selected teeth, is one of the most prevalent craniofacial anomalies in humans. Mutations in genes involved in WNT/β-catenin signaling, including AXIN2 WNT10A, WNT10B, LRP6, and KREMEN1, are known to cause FTA. However, mutational interactions among these genes have not been fully explored. In this study, we characterized four FTA kindreds with LRP6 pathogenic mutations: p.(Gln1252*), p.(Met168Arg), p.(Ala754Pro), and p.(Asn1075Ser). The three missense mutations were predicted to cause structural destabilization of the LRP6 protein. Two probands carrying both an LRP6 mutant allele and a WNT10A variant exhibited more severe phenotypes, suggesting mutational synergism or digenic inheritance. Biallelic LRP6 mutations in a patient with many missing teeth further supported the dose-dependence of LRP6-associated FTA. Analysis of 21 FTA cases with 15 different LRP6 loss-of-function mutations revealed high heterogeneity of disease severity and a distinctive pattern of missing teeth, with maxillary canines being frequently affected. We hypothesized that various combinations of sequence variants in WNT-related genes can modulate WNT signaling activities during tooth development and cause a wide spectrum of tooth agenesis severity, which highlights the importance of exome/genome analysis for the genetic diagnosis of FTA in this era of precision medicine.
Collapse
Affiliation(s)
- Kuan-Yu Chu
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei City 100, Taiwan; (K.-Y.C.); (Y.-L.W.); (J.-T.C.); (Y.-P.W.)
- Department of Pediatric Dentistry, National Taiwan University Children’s Hospital, Taipei City 100, Taiwan
| | - Yin-Lin Wang
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei City 100, Taiwan; (K.-Y.C.); (Y.-L.W.); (J.-T.C.); (Y.-P.W.)
- Department of Pediatric Dentistry, National Taiwan University Children’s Hospital, Taipei City 100, Taiwan
| | - Yu-Ren Chou
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei City 106, Taiwan;
| | - Jung-Tsu Chen
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei City 100, Taiwan; (K.-Y.C.); (Y.-L.W.); (J.-T.C.); (Y.-P.W.)
| | - Yi-Ping Wang
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei City 100, Taiwan; (K.-Y.C.); (Y.-L.W.); (J.-T.C.); (Y.-P.W.)
| | - James P. Simmer
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48108, USA; (J.P.S.); (J.C.-C.H.)
| | - Jan C.-C. Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48108, USA; (J.P.S.); (J.C.-C.H.)
| | - Shih-Kai Wang
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei City 100, Taiwan; (K.-Y.C.); (Y.-L.W.); (J.-T.C.); (Y.-P.W.)
- Department of Pediatric Dentistry, National Taiwan University Children’s Hospital, Taipei City 100, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 70251)
| |
Collapse
|
39
|
Rare phenotype: Hand preaxial polydactyly associated with LRP6-related tooth agenesis in humans. NPJ Genom Med 2021; 6:93. [PMID: 34759310 PMCID: PMC8581002 DOI: 10.1038/s41525-021-00262-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) is a pathogenic gene of selective tooth agenesis-7 (OMIM#616724). Although the malformation of the digits and fore- and hindlimbs has been reported in Lrp6-deficient mice, it has been rarely discovered in humans with LRP6 mutations. Here, we demonstrate an unreported autosomal dominant LRP6 heterozygous mutation (c.2840 T > C;p.Met947Thr) in a tooth agenesis family with hand polydactyly, and another unreported autosomal dominant LRP6 heterozygous mutation (c.1154 G > C;p.Arg385Pro) in a non-syndromic tooth agenesis family. Bioinformatic prediction demonstrated the deleterious effects of the mutations, and LRP6 structure changes suggested the corresponding functional impairments. Analysis on the pattern of LRP6-related tooth agenesis demonstrated the maxillary lateral incisor was the most affected. Our study report that LRP6 mutation might be associated with hand preaxial polydactyly in humans, which broaden the phenotypic spectrum of LRP6-related disorders, and provide valuable information on the characteristics of LRP6-related tooth agenesis.
Collapse
|
40
|
Identification of Rare LRP5 Variants in a Cohort of Males with Impaired Bone Mass. Int J Mol Sci 2021; 22:ijms221910834. [PMID: 34639175 PMCID: PMC8509722 DOI: 10.3390/ijms221910834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is the most common bone disease characterized by reduced bone mass and increased bone fragility. Genetic contribution is one of the main causes of primary osteoporosis; therefore, both genders are affected by this skeletal disorder. Nonetheless, osteoporosis in men has received little attention, thus being underestimated and undertreated. The aim of this study was to identify novel genetic variants in a cohort of 128 males with idiopathic low bone mass using a next-generation sequencing (NGS) panel including genes whose mutations could result in reduced bone mineral density (BMD). Genetic analysis detected in eleven patients ten rare heterozygous variants within the LRP5 gene, which were categorized as VUS (variant of uncertain significance), likely pathogenic and benign variants according to American College of Medical Genetics and Genomics (ACMG) guidelines. Protein structural and Bayesian analysis performed on identified LRP5 variants pointed out p.R1036Q and p.R1135C as pathogenic, therefore suggesting the likely association of these two variants with the low bone mass phenotype. In conclusion, this study expands our understanding on the importance of a functional LRP5 protein in bone formation and highlights the necessity to sequence this gene in subjects with idiopathic low BMD.
Collapse
|
41
|
Jeong W, Jho EH. Regulation of the Low-Density Lipoprotein Receptor-Related Protein LRP6 and Its Association With Disease: Wnt/β-Catenin Signaling and Beyond. Front Cell Dev Biol 2021; 9:714330. [PMID: 34589484 PMCID: PMC8473786 DOI: 10.3389/fcell.2021.714330] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
Wnt signaling plays crucial roles in development and tissue homeostasis, and its dysregulation leads to various diseases, notably cancer. Wnt/β-catenin signaling is initiated when the glycoprotein Wnt binds to and forms a ternary complex with the Frizzled and low-density lipoprotein receptor-related protein 5/6 (LRP5/6). Despite being identified as a Wnt co-receptor over 20 years ago, the molecular mechanisms governing how LRP6 senses Wnt and transduces downstream signaling cascades are still being deciphered. Due to its role as one of the main Wnt signaling components, the dysregulation or mutation of LRP6 is implicated in several diseases such as cancer, neurodegeneration, metabolic syndrome and skeletal disease. Herein, we will review how LRP6 is activated by Wnt stimulation and explore the various regulatory mechanisms involved. The participation of LRP6 in other signaling pathways will also be discussed. Finally, the relationship between LRP6 dysregulation and disease will be examined in detail.
Collapse
Affiliation(s)
- Wonyoung Jeong
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, South Korea
| |
Collapse
|
42
|
Jeong Y, Kim J, Choi HJ, Chung KY. Conformational Dynamics of Sclerostin-LRP6 Complex Analyzed by HDX-MS. Biomol Ther (Seoul) 2021; 29:527-535. [PMID: 33833136 PMCID: PMC8411024 DOI: 10.4062/biomolther.2020.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 11/26/2022] Open
Abstract
Sclerostin (SOST), a regulator of bone formation in osteocytes, inhibits the canonical Wnt signaling by interacting with low-density lipoprotein receptor-related protein 5/6 (LRP5/6) to prevent Wnt binding. Loss-of-function mutations of the SOST gene caused massive bone outgrowth and SOST-null mouse exhibited a high bone density phenotype. Therefore, SOST has been suggested as a promising therapeutic target for osteoporosis. A few previous studies with X-ray crystallography identified the binding interfaces between LRP6 and SOST, but there are limitations in these studies as they used truncated SOST protein or SOST peptide. Here, we analyzed the conformational dynamics of SOST-LRP6 E1E2 complex using hydrogen/deuterium exchange mass spectrometry (HDX-MS). We examined the effect of the C-terminal tail of SOST on LRP6 conformation upon complex formation. HDX-MS analysis suggested a new potential binding interface for the C-terminal region of SOST that was missing from the previous crystal structure of the SOST-LRP6 E1E2 complex.
Collapse
Affiliation(s)
- Yejing Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jinuk Kim
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee-Jung Choi
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
43
|
Bonnet C, Brahmbhatt A, Deng SX, Zheng JJ. Wnt signaling activation: targets and therapeutic opportunities for stem cell therapy and regenerative medicine. RSC Chem Biol 2021; 2:1144-1157. [PMID: 34458828 PMCID: PMC8341040 DOI: 10.1039/d1cb00063b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Wnt proteins are secreted morphogens that play critical roles in embryonic development, stem cell proliferation, self-renewal, tissue regeneration and remodeling in adults. While aberrant Wnt signaling contributes to diseases such as cancer, activation of Wnt/β-catenin signaling is a target of interest in stem cell therapy and regenerative medicine. Recent high throughput screenings from chemical and biological libraries, combined with improved gene expression reporter assays of Wnt/β-catenin activation together with rational drug design, led to the development of a myriad of Wnt activators, with different mechanisms of actions. Among them, Wnt mimics, antibodies targeting Wnt inhibitors, glycogen-synthase-3β inhibitors, and indirubins and other natural product derivatives are emerging modalities to treat bone, neurodegenerative, eye, and metabolic disorders, as well as prevent ageing. Nevertheless, the creation of Wnt-based therapies has been hampered by challenges in developing potent and selective Wnt activators without off-target effects, such as oncogenesis. On the other hand, to avoid these risks, their use to promote ex vivo expansion during tissue engineering is a promising application.
Collapse
Affiliation(s)
- Clémence Bonnet
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- INSERM, UMRS1138, Team 17, From Physiopathology of Ocular Diseases to Clinical Development, Paris University, Centre de Recherche des Cordeliers, and Cornea Departement, Cochin Hospital, AP-HP F-75014 Paris France
| | - Anvi Brahmbhatt
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
| | - Sophie X Deng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| | - Jie J Zheng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| |
Collapse
|
44
|
Ubels JL, Diegel CR, Foxa GE, Ethen NJ, Lensing JN, Madaj ZB, Williams BO. Low-Density Lipoprotein Receptor-Related Protein 5-Deficient Rats Have Reduced Bone Mass and Abnormal Development of the Retinal Vasculature. CRISPR J 2021; 3:284-298. [PMID: 32833527 DOI: 10.1089/crispr.2020.0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Humans carrying homozygous loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), develop osteoporosis and a defective retinal vasculature known as familial exudative vitreoretinopathy (FEVR) due to disruption of the Wnt signaling pathway. The purpose of this study was to use CRISPR-Cas9-mediated gene editing to create strains of Lrp5-deficient rats and to determine whether knockout of Lrp5 resulted in phenotypes that model the bone and retina pathology in LRP5-deficient humans. Knockout of Lrp5 in rats produced low bone mass, decreased bone mineral density, and decreased bone size. The superficial retinal vasculature of Lrp5-deficient rats was sparse and disorganized, with extensive exudates and decreases in vascularized area, vessel length, and branch point density. This study showed that Lrp5 could be predictably knocked out in rats using CRISPR-Cas9, causing the expression of bone and retinal phenotypes that will be useful for studying the role of Wnt signaling in bone and retina development and for research on the treatment of osteoporosis and FEVR.
Collapse
Affiliation(s)
- John L Ubels
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA.,Department of Biology, Calvin University, Grand Rapids, Michigan, USA
| | - Cassandra R Diegel
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA
| | - Gabrielle E Foxa
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA
| | - Nicole J Ethen
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA
| | - Jonathan N Lensing
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA
| | - Zachary B Madaj
- Core Technologies and Services, Van Andel Institute, Grand Rapids, Michigan, USA; Calvin University, Grand Rapids, Michigan, USA
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, and Calvin University, Grand Rapids, Michigan, USA
| |
Collapse
|
45
|
Manandhar S, Priya K, Mehta CH, Nayak UY, Kabekkodu SP, Pai KSR. Repositioning of antidiabetic drugs for Alzheimer's disease: possibility of Wnt signaling modulation by targeting LRP6 an in silico based study. J Biomol Struct Dyn 2021; 40:9577-9591. [PMID: 34080526 DOI: 10.1080/07391102.2021.1930583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022]
Abstract
Alzheimer disease (AD) is the most common, irreversible and progressive form of dementia for which the exact pathology and cause are still not clear. At present, we are only confined to symptomatic treatment, and the lack of disease-modifying therapeutics is worrisome. Alteration of Wnt signaling has been linked to metabolic diseases as well as AD. The crosstalk between Canonical Wnt signaling and insulin signaling pathway has been widely studied and accepted from several clinical and preclinical studies that have proven the beneficial effect of antidiabetic medications in the case of memory and cognition loss. This structure-based in silico study was focused on exploring the link between the currently available FDA approved antidiabetic drugs and the Wnt signaling pathway. The library of antidiabetics was obtained from drug bank and was screened for their binding affinity with protein (PDB ID: 3S2K) LRP6, a coreceptor of the Wnt signaling pathway using GLIDE module of Schrodinger. The top molecules, with higher docking score, binding energy and stable interactions, were subjected to energy-based calculation using MMGBSA, followed by a molecular dynamics-based simulation study. Drugs of class α-glucosidase inhibitors and peroxisome proliferator-activated receptors (PPARs) agonists were found to have a strong affinity towards LRP6 proteins, highlighting the possibility of the modulation of Wnt signaling by antidiabetics as one of the possible mechanisms for use in AD. However, further experimental based in vitro and in vivo studies are warranted for verification and support.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suman Manandhar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Keerthi Priya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
46
|
The structural biology of canonical Wnt signalling. Biochem Soc Trans 2021; 48:1765-1780. [PMID: 32725184 PMCID: PMC7458405 DOI: 10.1042/bst20200243] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022]
Abstract
The Wnt signalling pathways are of great importance in embryonic development and oncogenesis. Canonical and non-canonical Wnt signalling pathways are known, with the canonical (or β-catenin dependent) pathway being perhaps the best studied of these. While structural knowledge of proteins and interactions involved in canonical Wnt signalling has accumulated over the past 20 years, the pace of discovery has increased in recent years, with the structures of several key proteins and assemblies in the pathway being released. In this review, we provide a brief overview of canonical Wnt signalling, followed by a comprehensive overview of currently available X-ray, NMR and cryoEM data elaborating the structures of proteins and interactions involved in canonical Wnt signalling. While the volume of structures available is considerable, numerous gaps in knowledge remain, particularly a comprehensive understanding of the assembly of large multiprotein complexes mediating key aspects of pathway, as well as understanding the structure and activation of membrane receptors in the pathway. Nonetheless, the presently available data affords considerable opportunities for structure-based drug design efforts targeting canonical Wnt signalling.
Collapse
|
47
|
Bayle E, Svensson F, Atkinson BN, Steadman D, Willis NJ, Woodward HL, Whiting P, Vincent JP, Fish PV. Carboxylesterase Notum Is a Druggable Target to Modulate Wnt Signaling. J Med Chem 2021; 64:4289-4311. [PMID: 33783220 PMCID: PMC8172013 DOI: 10.1021/acs.jmedchem.0c01974] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Indexed: 12/12/2022]
Abstract
Regulation of the Wnt signaling pathway is critically important for a number of cellular processes in both development and adult mammalian biology. This Perspective will provide a summary of current and emerging therapeutic opportunities in modulating Wnt signaling, especially through inhibition of Notum carboxylesterase activity. Notum was recently shown to act as a negative regulator of Wnt signaling through the removal of an essential palmitoleate group. Inhibition of Notum activity may represent a new approach to treat disease where aberrant Notum activity has been identified as the underlying cause. Reliable screening technologies are available to identify inhibitors of Notum, and structural studies are accelerating the discovery of new inhibitors. A selection of these hits have been optimized to give fit-for-purpose small molecule inhibitors of Notum. Three noteworthy examples are LP-922056 (26), ABC99 (27), and ARUK3001185 (28), which are complementary chemical tools for exploring the role of Notum in Wnt signaling.
Collapse
Affiliation(s)
- Elliott
D. Bayle
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
- The
Francis Crick Institute, 1 Midland Road, Kings Cross, London NW1 1AT, U.K.
| | - Fredrik Svensson
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
- The
Francis Crick Institute, 1 Midland Road, Kings Cross, London NW1 1AT, U.K.
| | - Benjamin N. Atkinson
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
| | - David Steadman
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
| | - Nicky J. Willis
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
| | - Hannah L. Woodward
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
| | - Paul Whiting
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
| | - Jean-Paul Vincent
- The
Francis Crick Institute, 1 Midland Road, Kings Cross, London NW1 1AT, U.K.
| | - Paul V. Fish
- Alzheimer’s
Research UK UCL Drug Discovery Institute, University College London, Cruciform Building, Gower Street, London WC1E 6BT, U.K.
- The
Francis Crick Institute, 1 Midland Road, Kings Cross, London NW1 1AT, U.K.
| |
Collapse
|
48
|
Colozza G, Koo BK. Wnt/β-catenin signaling: Structure, assembly and endocytosis of the signalosome. Dev Growth Differ 2021; 63:199-218. [PMID: 33619734 PMCID: PMC8251975 DOI: 10.1111/dgd.12718] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022]
Abstract
Wnt/β‐catenin signaling is an ancient pathway that regulates key aspects of embryonic development, cell differentiation, proliferation, and adult stem cell homeostasis. Work from different laboratories has shed light on the molecular mechanisms underlying the Wnt pathway, including structural details of ligand–receptor interactions. One key aspect that has emerged from multiple studies is that endocytosis of the receptor complex plays a crucial role in fine‐tuning Wnt/β‐catenin signaling. Endocytosis is a key process involved in both activation as well as attenuation of Wnt signaling, but how this is regulated is still poorly understood. Importantly, recent findings show that Wnt also regulates central metabolic pathways such as the acquisition of nutrients through actin‐driven endocytic mechanisms. In this review, we propose that the Wnt pathway displays diverse characteristics that go beyond the regulation of gene expression, through a connection with the endocytic machinery.
Collapse
Affiliation(s)
- Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
49
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
50
|
Singh HD, Ma JX, Takahashi Y. Distinct roles of LRP5 and LRP6 in Wnt signaling regulation in the retina. Biochem Biophys Res Commun 2021; 545:8-13. [PMID: 33545636 DOI: 10.1016/j.bbrc.2021.01.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Dysregulation of Wnt signaling is implicated in multiple ocular disorders. The roles of Wnt co-receptors LRP5 and LRP6 in Wnt signaling regulation remain elusive, as most retinal cells express both of the co-receptors. To address this question, LRP5 and LRP6 were individually knocked-out in a human retinal pigment epithelium cell line using the CRISPR-Cas9 technology. Wnt signaling activity induced by various Wnt ligands was measured using wild-type and the KO cell lines. The results identified three groups of Wnt ligands based on their co-receptor specificity: 1) activation of Wnt signaling only through LRP6, 2) through both LRP5 and LRP6 and 3) predominantly through LRP5. These results indicate that LRP5 and LRP6 have differential roles in Wnt signaling regulation.
Collapse
Affiliation(s)
- Harminder D Singh
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yusuke Takahashi
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|