1
|
Chen Y, Bai Y, Deng D, Liu Y, He L, Wang M, Zhou M, Wang X, Yu C, Wang Y, Zhao F, Hai C, Wang B, Liu J, Kong X, Tu X, Li H. Novel LBR pathogenic variants with loss of sterol reductase activity participate in the pathogenesis of skeletal dysplasia via dysregulating canonical Wnt pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167901. [PMID: 40355051 DOI: 10.1016/j.bbadis.2025.167901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/15/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Biallelic pathogenic variants in the lamin B receptor (LBR) with impaired sterol reductase function are associated with the development of perinatal lethal Greenberg dysplasia (GRBGD) and mild nonfatal skeletal dysplasia with or without Pelger-Huet anomaly (PHASK), as well as other related hereditary skeletal dysplasia. However, the underlying molecular mechanism remains unclear. In this study, we found two novel pathogenic variants of LBR, namely missense mutation (c.1011 T > G, NM_002296.4; p.Cys337Trp, NP_002287.2) and LBR gene deletion (Chr1q42.12 (225,515,082-225,633,464), NC_000001.10). LBR is a novel substrate of FBW7, which is degraded by GSK3β/FBW7-mediated proteasome pathway and whose C337W mutation promotes its degradation through enhanced interaction with FBW7. Wild-type but not C337W mutant LBR is upregulated by WNT3A-mediated inactivation of GSK3β/FBW7 axis and then participated in WNT3A-activated Wnt pathway through its mediated cholesterol synthesis. MC3T3-E1 cells with Lbr knockdown or cholesterol removal exhibited reduced mineralized nodules in the presence of WNT3A, but addition of cholesterol in the culture medium reversed this phenotype. Collectively, we detected two novel variants in LBR and our study revealed for the first time that disruption of cholesterol synthesis by LBR impairs Wnt pathway and thus disrupts the cell osteogenic differentiation, providing new insights into the pathogenesis of skeletal dysplasia caused by LBR variation.
Collapse
Affiliation(s)
- Yilin Chen
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Ying Bai
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Dan Deng
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Yiheng Liu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Linyang He
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Mengru Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Mengchen Zhou
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xiangyi Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Chenguang Yu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Yue Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Feifei Zhao
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Chengying Hai
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Bo Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Junyi Liu
- Albany Medical College, New York 12208, USA
| | - Xiangdong Kong
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Xin Tu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| | - Hui Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| |
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
|
Sandhu K, Sahoo S, Arulandu A, Chockalingam S. Anaplastic lymphoma kinase enhances Wnt signaling through R-spondin: A new dimension to ALK-mediated oncogenesis. Int J Biol Macromol 2025; 308:142413. [PMID: 40132715 DOI: 10.1016/j.ijbiomac.2025.142413] [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: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Anaplastic lymphoma kinase receptor (ALK) is a receptor tyrosine kinase that plays a key role in the progression of several cancers and is activated by ligands such as ALKAL1 and ALKAL2. To identify additional molecules that interact with ALK, we constructed comprehensive genetic and molecular level networks. Notably, our study identified R-spondins, growth factors known to enhance Wnt signaling, as novel interacting partners of ALK. Protein-protein docking studies revealed that R-spondins bind to the TNF-like and EGF-like domains of ALK, which are critical for the interaction of ALK with its known ligand ALKAL2. These docking outcomes were further validated by molecular dynamics simulations, and approximate binding affinity calculations that confirmed the stability and conformational behavior of the ALK and R-spondin complex. These in silico findings indicate a strong interaction between ALK and R-spondins. To investigate whether this interaction influences Wnt signaling in vitro, we conducted a Wnt signaling reporter assay (TOP Flash/FOP Flash) in neuroblastoma cells by introducing Rspo2, Wnt3a, and crizotinib, an ALK inhibitor. The results showed a decrease in the TOP/FOP ratio when ALK was inhibited. Collectively, our study reveals a novel role for ALK in enhancing Wnt signaling via R-spondins, providing new dimension into ALK-mediated oncogenesis.
Collapse
Affiliation(s)
- Kajal Sandhu
- Cell Signaling Research Laboratory, Department of Biotechnology, National Institute of Technology Warangal, India
| | - Sibasis Sahoo
- Structural Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Arockiasamy Arulandu
- Structural Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - S Chockalingam
- Cell Signaling Research Laboratory, Department of Biotechnology, National Institute of Technology Warangal, India.
| |
Collapse
|
4
|
Chevallier L, Green M, Vo J, Vernau K, Marcellin-Little DJ, Jagannathan V, Leeb T, Bannasch D. The RSPO2 gene is associated with bilateral anterior amelia in Chihuahuas. Mamm Genome 2025:10.1007/s00335-025-10123-1. [PMID: 40131457 DOI: 10.1007/s00335-025-10123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025]
Abstract
Bilateral anterior amelia (BAA) is the congenital absence of thoracic limbs and has been reported in the Chihuahua as an autosomal recessive disorder. In some cases, the digits of the pelvic limbs can be variably affected, but otherwise, the pelvic limbs are generally spared. A GWAS performed with nine BAA affected Chihuahuas identified a significant association on chromosome 13, and homozygosity mapping delineated a 2.1 Mb chromosomal region containing the RSPO2 gene. Loss of function variants of RSPO2 in humans and cattle has been associated with the absence of all limbs. Six affected Chihuahuas were whole genome sequenced (WGS) and aligned to the CanFam4 assembly. SNVs, small indels, and structural variants within the critical interval that fitted a recessive model were investigated. Three SNVs (NC_049234.1:g.8891861C > T; NC_049234.1:g.8974204C > T and NC_049234.1:g.9789424G > A) were homozygous in five cases and absent from 3,418 genetically diverse control genome sequences, except for one Small Poodle that was heterozygous. One SNV resided in RSPO2's second intron, while the two others were intergenic. The three candidate variants were genotyped in 7 additional cases and 100 control Chihuahuas. Twelve of 13 cases were homozygous for the mutant allele, and one case was heterozygous. Controls were either homozygous for the reference allele (97%) or heterozygous (3%). Our data should facilitate genetic testing of Chihuahuas to prevent the unintentional production of BAA affected dogs. Moreover, the identification of these variants enhances understanding of RSPO2 gene function in limb development.
Collapse
Affiliation(s)
- Lucie Chevallier
- INSERM, UPEC, Ecole Nationale Vétérinaire d'Alfort, U955 - IMRB, Team 10 - Biology of the Neuromuscular System, Maisons-Alfort, France
| | - Marin Green
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - Julia Vo
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - Karen Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - Denis J Marcellin-Little
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland
| | - Danika Bannasch
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA.
| |
Collapse
|
5
|
Lim W. LGR4 (GPR48): The Emerging Inter-Bridge in Osteoimmunology. Biomedicines 2025; 13:607. [PMID: 40149584 PMCID: PMC11940432 DOI: 10.3390/biomedicines13030607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), a member of the G-protein-coupled receptor (GPCR) family, has been implicated in various regulatory functions across multiple differentiation stages and numerous target sites in bone diseases. Therefore, LGR4 is a potential regulator of nuclear factor-κB ligand (RANKL) during osteoclast differentiation. However, a comprehensive investigation of its functions and applications in bone immunology is lacking. This review discusses the molecular characteristics, signaling pathways, and role of LGR4 in osteoimmunology, with a particular focus on its interactions with RANKL during osteoclast differentiation, while identifying gaps that warrant further research.
Collapse
Affiliation(s)
- Wonbong Lim
- Department of Orthopaedic Surgery, Chosun University, Gwangju 61453, Republic of Korea; ; Tel.: +82-62-230-6193; Fax: +82-62-226-3379
- Laboratory of Orthopaedic Research, Chosun University, Gwangju 61453, Republic of Korea
- Regional Leading Research Center, Chonnam National University, Yeosu 59626, Republic of Korea
- Department of Premedical Program, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
6
|
Yue F, Ku AT, Stevens PD, Michalski MN, Jiang W, Tu J, Shi Z, Dou Y, Wang Y, Feng XH, Hostetter G, Wu X, Huang S, Shroyer NF, Zhang B, Williams BO, Liu Q, Lin X, Li Y. Loss of ZNRF3/RNF43 Unleashes EGFR in Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574969. [PMID: 38260423 PMCID: PMC10802575 DOI: 10.1101/2024.01.10.574969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
ZNRF3 and RNF43 are closely related transmembrane E3 ubiquitin ligases with significant roles in development and cancer. Conventionally, their biological functions have been associated with regulating WNT signaling receptor ubiquitination and degradation. However, our proteogenomic studies have revealed EGFR as the protein most negatively correlated with ZNRF3/RNF43 mRNA levels in multiple human cancers. Through biochemical investigations, we demonstrate that ZNRF3/RNF43 interact with EGFR via their extracellular domains, leading to EGFR ubiquitination and subsequent degradation facilitated by the E3 ligase RING domain. Overexpression of ZNRF3 reduces EGFR levels and suppresses cancer cell growth in vitro and in vivo, whereas knockout of ZNRF3/RNF43 stimulates cell growth and tumorigenesis through upregulated EGFR signaling. Together, these data highlight ZNRF3 and RNF43 as novel E3 ubiquitin ligases of EGFR and establish the inactivation of ZNRF3/RNF43 as a driver of increased EGFR signaling, ultimately promoting cancer progression. This discovery establishes a connection between two fundamental signaling pathways, EGFR and WNT, at the level of cytoplasmic membrane receptors, uncovering a novel mechanism underlying the frequent co-activation of EGFR and WNT signaling in development and cancer.
Collapse
Affiliation(s)
- Fei Yue
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Amy T. Ku
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Payton D. Stevens
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
- Biological Sciences Department, Miami University, Oxford, Ohio, 45056, USA
| | - Megan N. Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
| | - Weiyu Jiang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jianghua Tu
- Texas Therapeutics Institute and Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Zhongcheng Shi
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Galen Hostetter
- Van Andel Institute, Core Technologies and Services, Grand Rapids, Michigan 49503, USA
| | - Xiangwei Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shixia Huang
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Education, Innovation & Technology, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Noah F. Shroyer
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Bart O. Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
- Van Andel Institute, Core Technologies and Services, Grand Rapids, Michigan 49503, USA
| | - Qingyun Liu
- Texas Therapeutics Institute and Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Xia Lin
- The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
7
|
Luo D, Zheng J, Lv S, Sheng R, Chen M, He X, Zhang X. Wnt specifically induces FZD5/8 endocytosis and degradation and the involvement of RSPO-ZNRF3/RNF43 and DVL. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619000. [PMID: 39463927 PMCID: PMC11507892 DOI: 10.1101/2024.10.18.619000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Frizzled (FZD) proteins are the principal receptors of the Wnt signaling pathway. However, whether Wnt ligands induce FZD endocytosis and degradation remains elusive. The transmembrane E3 ubiquitin ligases ZNRF3 and RNF43 promote the endocytosis and degradation of FZD receptors to inhibit Wnt signaling, and their function is antagonized by R-spondin (RSPO) proteins. However, the dependency of RSPO-ZNRF3/RNF43-mediated FZD endocytosis and degradation on Wnt stimulation, as well as the specificity of this degradation for different FZD, remains unclear. Here, we demonstrated that Wnt specifically induces FZD5/8 endocytosis and degradation in a ZNRF3/RNF43-dependent manner. ZNRF3/RNF43 selectively targets FZD5/8 for degradation upon Wnt stimulation. RSPO1 enhances Wnt signaling by specifically stabilizing FZD5/8. Wnt promotes the interaction between FZD5 and RNF43. We further demonstrated that DVL proteins promote ligand-independent endocytosis of FZD but are dispensable for Wnt-induced FZD5/8 endocytosis and degradation. Our results reveal a novel negative regulatory mechanism of Wnt signaling at the receptor level and illuminate the mechanism by which RSPO-ZNRF3/RNF43 regulates Wnt signaling, which may provide new insights into regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Dong Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Jing Zheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Shuning Lv
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ren Sheng
- College of Life and Health Science, Northeastern University, Shenyang, China
| | - Maorong Chen
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xi He
- The F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Xinjun Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
8
|
Zhang S, Zhu J, Jin S, Sun W, Ji W, Chen Z. Jawbone periosteum-derived cells with high osteogenic potential controlled by R-spondin 3. FASEB J 2024; 38:e70079. [PMID: 39340242 DOI: 10.1096/fj.202400988rr] [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: 05/02/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
The jawbone periosteum, the easily accessible tissue responding to bone repair, has been overlooked in the recent development of cell therapy for jawbone defect reconstruction. Therefore, this study aimed to elucidate the in vitro and in vivo biological characteristics of jawbone periosteum-derived cells (jb-PDCs). For this purpose, we harvested the jb-PDCs from 8-week-old C57BL/6 mice. The in vitro cultured jb-PDCs (passages 1 and 3) contained skeletal stem/progenitor cells and exhibited clonogenicity and tri-lineage differentiation capacity. When implanted in vivo, the jb-PDCs (passage 3) showed evident ectopic bone formation after 4-week subcutaneous implantation, and active contribution to repair the critical-size jawbone defects in mice. Molecular profiling suggested that R-spondin 3 was strongly associated with the superior in vitro and in vivo osteogenic potentials of jb-PDCs. Overall, our study highlights the significance of comprehending the biological characteristics of the jawbone periosteum, which could pave the way for innovative cell-based therapies for the reconstruction of jawbone defects.
Collapse
Affiliation(s)
- Shu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingxian Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Siyu Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Sun
- Department of Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
King JS, Wan M, Wagley Y, Stestiv M, Kalajzic I, Hankenson KD, Sanjay A. Signaling pathways associated with Lgr6 to regulate osteogenesis. Bone 2024; 187:117207. [PMID: 39033993 DOI: 10.1016/j.bone.2024.117207] [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: 05/10/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Fracture management largely relies on the bone's inherent healing capabilities and, when necessary, surgical intervention. Currently, there are limited osteoinductive therapies to promote healing, making targeting skeletal stem/progenitor cells (SSPCs) a promising avenue for therapeutic development. A limiting factor for this approach is our incomplete understanding of the molecular mechanisms governing SSPCs' behavior. We have recently identified that the Leucine-rich repeat-containing G-protein coupled receptor 6 (Lgr6) is expressed in sub-populations of SSPCs, and is required for maintaining bone volume during adulthood and for proper fracture healing. Lgr family members (Lgr4-6) are markers of stem cell niches and play a role in tissue regeneration primarily by binding R-Spondin (Rspo1-4). This interaction promotes canonical Wnt (cWnt) signaling by stabilizing Frizzled receptors. Interestingly, our findings here indicate that Lgr6 may also influence cWnt-independent pathways. Remarkably, Lgr6 expression was enhanced during Bmp-mediated osteogenesis of both human and murine cells. Using biochemical approaches, RNA sequencing, and bioinformatic analysis of published single-cell data, we found that elements of BMP signaling, including its target gene, pSMAD, and gene ontology pathways, are downregulated in the absence of Lgr6. Our findings uncover a molecular interdependency between the Bmp pathway and Lgr6, offering new insights into osteogenesis and potential targets for enhancing fracture healing.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Marta Stestiv
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA.
| |
Collapse
|
10
|
Niehrs C, Seidl C, Lee H. An "R-spondin code" for multimodal signaling ON-OFF states. Bioessays 2024; 46:e2400144. [PMID: 39180250 DOI: 10.1002/bies.202400144] [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: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
R-spondins (RSPOs) are a family of secreted proteins and stem cell growth factors that are potent co-activators of Wnt signaling. Recently, RSPO2 and RSPO3 were shown to be multifunctional, not only amplifying Wnt- but also binding BMP- and FGF receptors to downregulate signaling. The common mechanism underlying these diverse functions is that RSPO2 and RSPO3 act as "endocytosers" that link transmembrane proteins to ZNRF3/RNF43 E3 ligases and trigger target internalization. Thus, RSPOs are natural protein targeting chimeras for cell surface proteins. Conducting data mining and cell surface binding assays we report additional candidate RSPO targets, including SMO, PTC1,2, LGI1, ROBO4, and PTPR(F/S). We propose that there is an "R-spondin code" that imparts combinatorial signaling ON-OFF states of multiple growth factors. This code involves the modular RSPO domains, notably distinct motifs in the divergent RSPO-TSP1 domains to mediate target interaction and internalization. The RSPO code offers a novel framework for the understanding how diverse signaling pathways may be coordinately regulated in development and disease.
Collapse
Affiliation(s)
- Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Carina Seidl
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Hyeyoon Lee
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Boonsawat P, Asadollahi R, Niedrist D, Steindl K, Begemann A, Joset P, Bhoj EJ, Li D, Zackai E, Vetro A, Barba C, Guerrini R, Whalen S, Keren B, Khan A, Jing D, Palomares Bralo M, Rikeros Orozco E, Hao Q, Schlott Kristiansen B, Zheng B, Donnelly D, Clowes V, Zweier M, Papik M, Siegel G, Sabatino V, Mocera M, Horn AHC, Sticht H, Rauch A. Deleterious ZNRF3 germline variants cause neurodevelopmental disorders with mirror brain phenotypes via domain-specific effects on Wnt/β-catenin signaling. Am J Hum Genet 2024; 111:1994-2011. [PMID: 39168120 PMCID: PMC11393693 DOI: 10.1016/j.ajhg.2024.07.016] [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: 04/29/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Zinc and RING finger 3 (ZNRF3) is a negative-feedback regulator of Wnt/β-catenin signaling, which plays an important role in human brain development. Although somatically frequently mutated in cancer, germline variants in ZNRF3 have not been established as causative for neurodevelopmental disorders (NDDs). We identified 12 individuals with ZNRF3 variants and various phenotypes via GeneMatcher/Decipher and evaluated genotype-phenotype correlation. We performed structural modeling and representative deleterious and control variants were assessed using in vitro transcriptional reporter assays with and without Wnt-ligand Wnt3a and/or Wnt-potentiator R-spondin (RSPO). Eight individuals harbored de novo missense variants and presented with NDD. We found missense variants associated with macrocephalic NDD to cluster in the RING ligase domain. Structural modeling predicted disruption of the ubiquitin ligase function likely compromising Wnt receptor turnover. Accordingly, the functional assays showed enhanced Wnt/β-catenin signaling for these variants in a dominant negative manner. Contrarily, an individual with microcephalic NDD harbored a missense variant in the RSPO-binding domain predicted to disrupt binding affinity to RSPO and showed attenuated Wnt/β-catenin signaling in the same assays. Additionally, four individuals harbored de novo truncating or de novo or inherited large in-frame deletion variants with non-NDD phenotypes, including heart, adrenal, or nephrotic problems. In contrast to NDD-associated missense variants, the effects on Wnt/β-catenin signaling were comparable between the truncating variant and the empty vector and between benign variants and the wild type. In summary, we provide evidence for mirror brain size phenotypes caused by distinct pathomechanisms in Wnt/β-catenin signaling through protein domain-specific deleterious ZNRF3 germline missense variants.
Collapse
Affiliation(s)
| | - Reza Asadollahi
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Faculty of Engineering and Science, University of Greenwich London, Medway Campus, Chatham Maritime ME4 4TB, UK
| | - Dunja Niedrist
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anaïs Begemann
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Pascal Joset
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Elizabeth J Bhoj
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Carmen Barba
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Sandra Whalen
- Unité Fonctionnelle de Génétique Odellin, Hôpital Armand Trousseau, Paris, France
| | - Boris Keren
- Département de Génétique, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Amjad Khan
- Faculty of Science, Department of Biological Science (Zoology), University of Lakki Marwat, Khyber Pakhtunkhwa 28420, Pakistan
| | - Duan Jing
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - María Palomares Bralo
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Emi Rikeros Orozco
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Qin Hao
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Deirdre Donnelly
- Northern Ireland Regional Genetics Centre, Belfast Health & Social Care Trust, Belfast, Northern Ireland
| | - Virginia Clowes
- Thames Regional Genetics Service, North West University Healthcare NHS Trust, London, UK
| | - Markus Zweier
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Michael Papik
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Gabriele Siegel
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Valeria Sabatino
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Martina Mocera
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anselm H C Horn
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Pediatric University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
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
|
13
|
Niehrs C, Zapparoli E, Lee H. 'Three signals - three body axes' as patterning principle in bilaterians. Cells Dev 2024:203944. [PMID: 39121910 DOI: 10.1016/j.cdev.2024.203944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
In vertebrates, the three orthogonal body axes, anteroposterior (AP), dorsoventral (DV) and left-right (LR) are determined at gastrula and neurula stages by the Spemann-Mangold organizer and its equivalents. A common feature of AP and DV axis formation is that an evolutionary conserved interplay between growth factors (Wnt, BMP) and their extracellular antagonists (e.g. Dkk1, Chordin) creates signaling gradients for axial patterning. Recent work showed that LR patterning in Xenopus follows the same principle, with R-spondin 2 (Rspo2) as an extracellular FGF antagonist, which creates a signaling gradient that determines the LR vector. That a triad of anti-FGF, anti-BMP, and anti-Wnt governs LR, DV, and AP axis formation reveals a unifying principle in animal development. We discuss how cross-talk between these three signals confers integrated AP-DV-LR body axis patterning underlying developmental robustness, size scaling, and harmonious regulation. We propose that Urbilateria featured three orthogonal body axes that were governed by a Cartesian coordinate system of orthogonal Wnt/AP, BMP/DV, and FGF/LR signaling gradients.
Collapse
Affiliation(s)
- Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany; Institute of Molecular Biology (IMB), 55128 Mainz, Germany.
| | | | - Hyeyoon Lee
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
14
|
Liu Y, Fan M, Yang J, Mihaljević L, Chen KH, Ye Y, Sun S, Qiu Z. KAT6A deficiency impairs cognitive functions through suppressing RSPO2/Wnt signaling in hippocampal CA3. SCIENCE ADVANCES 2024; 10:eadm9326. [PMID: 38758792 PMCID: PMC11100567 DOI: 10.1126/sciadv.adm9326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Intellectual disability (ID) affects ~2% of the population and ID-associated genes are enriched for epigenetic factors, including those encoding the largest family of histone lysine acetyltransferases (KAT5-KAT8). Among them is KAT6A, whose mutations cause KAT6A syndrome, with ID as a common clinical feature. However, the underlying molecular mechanism remains unknown. Here, we find that KAT6A deficiency impairs synaptic structure and plasticity in hippocampal CA3, but not in CA1 region, resulting in memory deficits in mice. We further identify a CA3-enriched gene Rspo2, encoding Wnt activator R-spondin 2, as a key transcriptional target of KAT6A. Deletion of Rspo2 in excitatory neurons impairs memory formation, and restoring RSPO2 expression in CA3 neurons rescues the deficits in Wnt signaling and learning-associated behaviors in Kat6a mutant mice. Collectively, our results demonstrate that KAT6A-RSPO2-Wnt signaling plays a critical role in regulating hippocampal CA3 synaptic plasticity and cognitive function, providing potential therapeutic targets for KAT6A syndrome and related neurodevelopmental diseases.
Collapse
Affiliation(s)
- Yongqing Liu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Minghua Fan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Junhua Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ljubica Mihaljević
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kevin Hong Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yingzhi Ye
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Srivastava A, Rikhari D, Srivastava S. RSPO2 as Wnt signaling enabler: Important roles in cancer development and therapeutic opportunities. Genes Dis 2024; 11:788-806. [PMID: 37692504 PMCID: PMC10491879 DOI: 10.1016/j.gendis.2023.01.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 01/16/2023] [Indexed: 09/12/2023] Open
Abstract
R-spondins are secretory proteins localized in the endoplasmic reticulum and Golgi bodies and are processed through the secretory pathway. Among the R-spondin family, RSPO2 has emanated as a novel regulator of Wnt signaling, which has now been acknowledged in numerous in vitro and in vivo studies. Cancer is an abnormal growth of cells that proliferates and spreads uncontrollably due to the accumulation of genetic and epigenetic factors that constitutively activate Wnt signaling in various types of cancer. Colorectal cancer (CRC) begins when cells in the colon and rectum follow an indefinite pattern of division due to aberrant Wnt activation as one of the key hallmarks. Decades-long progress in research on R-spondins has demonstrated their oncogenic function in distinct cancer types, particularly CRC. As a critical regulator of the Wnt pathway, it modulates several phenotypes of cells, such as cell proliferation, invasion, migration, and cancer stem cell properties. Recently, RSPO mutations, gene rearrangements, fusions, copy number alterations, and altered gene expression have also been identified in a variety of cancers, including CRC. In this review, we addressed the recent updates regarding the recurrently altered R-spondins with special emphasis on the RSPO2 gene and its involvement in potentiating Wnt signaling in CRC. In addition to the compelling physiological and biological roles in cellular fate and regulation, we propose that RSPO2 would be valuable as a potential biomarker for prognostic, diagnostic, and therapeutic use in CRC.
Collapse
Affiliation(s)
- Ankit Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, Uttar Pradesh 211004, India
| | - Deeksha Rikhari
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, Uttar Pradesh 211004, India
| | - Sameer Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, Uttar Pradesh 211004, India
| |
Collapse
|
16
|
Srivastava A, Srivastava S. Multiomics data identifies RSPO2 as a prognostic biomarker in human tumors associated with pan-cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:469-499. [PMID: 38448143 DOI: 10.1016/bs.apcsb.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
RSPO2 protein may provide valuable insights into the mechanism underlying various types of tumorigenesis. The role of RSPO2 in pan-cancer has not been reported so far. Therefore, this study aimed to provide a comprehensive analysis of RSPO2 from a pan-cancer perspective employing multiomics data. The expression profile and function of RSPO2 across different tumors were investigated using various web-based tools UALCAN, GEPIA, TIMER, Human Protein Atlas, cBioPortal, TISIDB, STRING, and Metascape to interpret the expression profile, promoter methylation status, genomic alterations, survival analysis, protein-protein interaction, correlation with immune cell subtypes, tumor immune microenvironment and enrichment analysis. Comprehensive pan-cancer analysis indicated that RSPO2 was significantly downregulated in eleven and upregulated in five tumor types compared to normal tissues, validation results further suggest RSPO2 was downregulated in most of the tumors. The protein level expression of RSPO2 was mostly low in malignant tissues. We found that RSPO2 was significantly related to individual pathological stages in BLCA, COAD, LUAD and LUSC. Prognostic analysis indicates that the high RSPO2 expression was significantly correlated with the poor prognosis in BRCA, KICH, KIRP, READ, and UCES. Furthermore, RSPO2 is frequently amplified, exhibits hypermethylated promoter in most cancers, and is associated with immune subtypes, molecular subtypes and immune cell infiltration. Finally, enrichment analysis showed that RSPO2 is involved in the regulation of the canonical Wnt pathway and neuronal development. The overall comprehensive pan-cancer analysis affirms that RSPO2 could be a promising diagnostic and prognostic biomarker and latent therapy target in the future.
Collapse
Affiliation(s)
- Ankit Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, Uttar Pradesh, India
| | - Sameer Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, Uttar Pradesh, India.
| |
Collapse
|
17
|
Lee H, Camuto CM, Niehrs C. R-Spondin 2 governs Xenopus left-right body axis formation by establishing an FGF signaling gradient. Nat Commun 2024; 15:1003. [PMID: 38307837 PMCID: PMC10837206 DOI: 10.1038/s41467-024-44951-7] [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: 06/13/2023] [Accepted: 01/10/2024] [Indexed: 02/04/2024] Open
Abstract
Establishment of the left-right (LR, sinistral, dextral) body axis in many vertebrate embryos relies on cilia-driven leftward fluid flow within an LR organizer (LRO). A cardinal question is how leftward flow triggers symmetry breakage. The chemosensation model posits that ciliary flow enriches a signaling molecule on the left side of the LRO that promotes sinistral cell fate. However, the nature of this sinistralizing signal has remained elusive. In the Xenopus LRO, we identified the stem cell growth factor R-Spondin 2 (Rspo2) as a symmetrically expressed, sinistralizing signal. As predicted for a flow-mediated signal, Rspo2 operates downstream of leftward flow but upstream of the asymmetrically expressed gene dand5. Unexpectedly, in LR patterning, Rspo2 acts as an FGF receptor antagonist: Rspo2 via its TSP1 domain binds Fgfr4 and promotes its membrane clearance by Znrf3-mediated endocytosis. Concordantly, we find that at flow-stage, FGF signaling is dextralizing and forms a gradient across the LRO, high on the dextral- and low on the sinistral side. Rspo2 gain- and loss-of function equalize this FGF signaling gradient and sinistralize and dextralize development, respectively. We propose that leftward flow of Rspo2 produces an FGF signaling gradient that governs LR-symmetry breakage.
Collapse
Affiliation(s)
- Hyeyoon Lee
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120, Heidelberg, Germany
| | - Celine Marie Camuto
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120, Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120, Heidelberg, Germany.
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany.
| |
Collapse
|
18
|
Farnhammer F, Colozza G, Kim J. RNF43 and ZNRF3 in Wnt Signaling - A Master Regulator at the Membrane. Int J Stem Cells 2023; 16:376-384. [PMID: 37643759 PMCID: PMC10686798 DOI: 10.15283/ijsc23070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/10/2023] [Indexed: 08/31/2023] Open
Abstract
The Wnt β-catenin signaling pathway is a highly conserved mechanism that plays a critical role from embryonic development and adult stem cell homeostasis. However, dysregulation of the Wnt pathway has been implicated in various diseases, including cancer. Therefore, multiple layers of regulatory mechanisms tightly control the activation and suppression of the Wnt signal. The E3 ubiquitin ligases RNF43 and ZNRF3, which are known negative regulators of the Wnt pathway, are critical component of Wnt signaling regulation. These E3 ubiquitin ligases control Wnt signaling by targeting the Wnt receptor Frizzled to induce ubiquitination-mediated endo-lysosomal degradation, thus controlling the activation of the Wnt signaling pathway. We also discuss the regulatory mechanisms, interactors, and evolution of RNF43 and ZNRF3. This review article summarizes recent findings on RNF43 and ZNRF3 and their potential implications for the development of therapeutic strategies to target the Wnt signaling pathway in various diseases, including cancer.
Collapse
Affiliation(s)
- Fiona Farnhammer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
- Division of Oncology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Jihoon Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Korea
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Korea
| |
Collapse
|
19
|
Zhang L, Adu IK, Zhang H, Wang J. The WNT/β-catenin system in chronic kidney disease-mineral bone disorder syndrome. Int Urol Nephrol 2023; 55:2527-2538. [PMID: 36964322 DOI: 10.1007/s11255-023-03569-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/18/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND The WNT/β-catenin system is an evolutionarily conserved signaling pathway that plays a crucial role in morphogenesis and cell tissue formation during embryogenesis. Although usually suppressed in adulthood, it can be reactivated during organ damage and regeneration. Transient activation of the WNT/β-catenin pathway stimulates tissue regeneration after acute kidney injury, while persistent (uncontrolled) activation can promote the development of chronic kidney disease (CKD). CKD-MBD is a clinical syndrome that develops with systemic mineral and bone metabolism disorders caused by CKD, characterized by abnormal bone mineral metabolism and/or extraosseous calcification, as well as cardiovascular disease associated with CKD, including vascular stiffness and calcification. OBJECTIVE This paper aims to comprehensively review the WNT/β-catenin signaling pathway in relation to CKD-MBD, focusing on its components, regulatory molecules, and regulatory mechanisms. Additionally, this review highlights the challenges and opportunities for using small molecular compounds to target the WNT/β-catenin signaling pathway in CKD-MBD therapy. METHODS We conducted a comprehensive literature review using various scientific databases, including PubMed, Scopus, and Web of Science, to identify relevant articles. We searched for articles that discussed the WNT/β-catenin signaling pathway, CKD-MBD, and their relationship. We also reviewed articles that discussed the components of the WNT/β-catenin signaling pathway, its regulatory molecules, and regulatory mechanisms. RESULTS The WNT/β-catenin signaling pathway plays a crucial role in CKD-MBD by promoting vascular calcification and bone mineral metabolism disorders. The pathway's components include WNT ligands, Frizzled receptors, and LRP5/6 co-receptors, which initiate downstream signaling cascades leading to the activation of β-catenin. Several regulatory molecules, including GSK-3β, APC, and Axin, modulate β-catenin activation. The WNT/β-catenin signaling pathway also interacts with other signaling pathways, such as the BMP pathway, to regulate CKD-MBD. CONCLUSIONS The WNT/β-catenin signaling pathway is a potential therapeutic target for CKD-MBD. Small molecular compounds that target the components or regulatory molecules of the pathway may provide a promising approach to treat CKD-MBD. However, more research is needed to identify safe and effective compounds and to determine the optimal dosages and treatment regimens.
Collapse
Affiliation(s)
- Lingbo Zhang
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, People's Republic of China
| | - Isaac Kumi Adu
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, People's Republic of China
- Department of Internal Medicine, The Second Hospital of Jingzhou and the Affiliated Hospital of Hubei College of Chinese Medicine, Jingzhou, People's Republic of China
- Department of Internal Medicine, Kings and Queens University College and Teaching Hospital, Akosombo, Ghana
| | - Haifeng Zhang
- Department of Internal Medicine, The Second Hospital of Jingzhou and the Affiliated Hospital of Hubei College of Chinese Medicine, Jingzhou, People's Republic of China
| | - Jiancheng Wang
- Department of Internal Medicine, The Second Hospital of Jingzhou and the Affiliated Hospital of Hubei College of Chinese Medicine, Jingzhou, People's Republic of China.
| |
Collapse
|
20
|
Hossain N, Igawa T, Suzuki M, Tazawa I, Nakao Y, Hayashi T, Suzuki N, Ogino H. Phenotype-genotype relationships in Xenopus sox9 crispants provide insights into campomelic dysplasia and vertebrate jaw evolution. Dev Growth Differ 2023; 65:481-497. [PMID: 37505799 DOI: 10.1111/dgd.12884] [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: 05/18/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Since CRISPR-based genome editing technology works effectively in the diploid frog Xenopus tropicalis, a growing number of studies have successfully modeled human genetic diseases in this species. However, most of their targets were limited to non-syndromic diseases that exhibit abnormalities in a small fraction of tissues or organs in the body. This is likely because of the complexity of interpreting the phenotypic variations resulting from somatic mosaic mutations generated in the founder animals (crispants). In this study, we attempted to model the syndromic disease campomelic dysplasia (CD) by generating sox9 crispants in X. tropicalis. The resulting crispants failed to form neural crest cells at neurula stages and exhibited various combinations of jaw, gill, ear, heart, and gut defects at tadpole stages, recapitulating part of the syndromic phenotype of CD patients. Genotyping of the crispants with a variety of allelic series of mutations suggested that the heart and gut defects depend primarily on frame-shift mutations expected to be null, whereas the jaw, gill, and ear defects could be induced not only by such mutations but also by in-frame deletion mutations expected to delete part of the jawed vertebrate-specific domain from the encoded Sox9 protein. These results demonstrate that Xenopus crispants are useful for investigating the phenotype-genotype relationships behind syndromic diseases and examining the tissue-specific role of each functional domain within a single protein, providing novel insights into vertebrate jaw evolution.
Collapse
Affiliation(s)
- Nusrat Hossain
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Takeshi Igawa
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Makoto Suzuki
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Ichiro Tazawa
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Yuta Nakao
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Toshinori Hayashi
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Nanoka Suzuki
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
| | - Hajime Ogino
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
21
|
Post Y, Dilip A, Xie L, Sura A, Suen N, Ye J, Mutha D, Liu AT, Nguyen H, Whisler E, Shah D, Deshmukh S, Dhaliwal N, Bauer B, Nigatu E, Diep A, Lopez T, Fowler TW, Lee SJ, Lu C, Yeh WC, Chen H, Li Y. Novel Frizzled-specific antibody-based Wnt mimetics and Wnt superagonists selectively activate WNT/β-catenin signaling in target tissues. Cell Chem Biol 2023; 30:976-986.e5. [PMID: 37413985 DOI: 10.1016/j.chembiol.2023.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/18/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
WNTs are essential factors for stem cell biology, embryonic development, and for maintaining homeostasis and tissue repair in adults. Difficulties in purifying WNTs and their lack of receptor selectivity have hampered research and regenerative medicine development. While breakthroughs in WNT mimetic development have overcome some of these difficulties, the tools developed so far are incomplete and mimetics alone are often not sufficient. Here, we developed a complete set of WNT mimetic molecules that cover all WNT/β-catenin-activating Frizzleds (FZDs). We show that FZD1,2,7 stimulate salivary gland expansion in vivo and salivary gland organoid expansion. We further describe the discovery of a novel WNT-modulating platform that combines WNT and RSPO mimetics' effects into one molecule. This set of molecules supports better organoid expansion in various tissues. These WNT-activating platforms can be broadly applied to organoids, pluripotent stem cells, and in vivo research, and serve as bases for future therapeutic development.
Collapse
Affiliation(s)
- Yorick Post
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Archana Dilip
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Liqin Xie
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Asmiti Sura
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Nicholas Suen
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Jay Ye
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Devin Mutha
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Angela T Liu
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Huy Nguyen
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Elizabeth Whisler
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Darshini Shah
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Shalaka Deshmukh
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Navrose Dhaliwal
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Ben Bauer
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Eskedar Nigatu
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Anh Diep
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Tom Lopez
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Tristan W Fowler
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Sung-Jin Lee
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Chenggang Lu
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Wen-Chen Yeh
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Hui Chen
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA
| | - Yang Li
- Surrozen Inc., 171 Oyster Point Boulevard, Suite 400, South San Francisco, CA 94080, USA.
| |
Collapse
|
22
|
Fu XX, Zhuo DH, Zhang YJ, Li YF, Liu X, Xing YY, Huang Y, Wang YF, Cheng T, Wang D, Chen SH, Chen YJ, Jiang GN, Lu FI, Feng Y, Huang X, Ma J, Liu W, Bai G, Xu PF. A spatiotemporal barrier formed by Follistatin is required for left-right patterning. Proc Natl Acad Sci U S A 2023; 120:e2219649120. [PMID: 37276408 PMCID: PMC10268237 DOI: 10.1073/pnas.2219649120] [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: 11/18/2022] [Accepted: 04/25/2023] [Indexed: 06/07/2023] Open
Abstract
How left-right (LR) asymmetry emerges in a patterning field along the anterior-posterior axis remains an unresolved problem in developmental biology. Left-biased Nodal emanating from the LR organizer propagates from posterior to anterior (PA) and establishes the LR pattern of the whole embryo. However, little is known about the regulatory mechanism of the PA spread of Nodal and its asymmetric activation in the forebrain. Here, we identify bilaterally expressed Follistatin (Fst) as a regulator blocking the propagation of the zebrafish Nodal ortholog Southpaw (Spaw) in the right lateral plate mesoderm (LPM), and restricting Spaw transmission in the left LPM to facilitate the establishment of a robust LR asymmetric Nodal patterning. In addition, Fst inhibits the Activin-Nodal signaling pathway in the forebrain thus preventing Nodal activation prior to the arrival, at a later time, of Spaw emanating from the left LPM. This contributes to the orderly propagation of asymmetric Nodal activation along the PA axis. The LR regulation function of Fst is further confirmed in chick and frog embryos. Overall, our results suggest that a robust LR patterning emerges by counteracting a Fst barrier formed along the PA axis.
Collapse
Affiliation(s)
- Xin-Xin Fu
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Ding-Hao Zhuo
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Ying-Jie Zhang
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Yun-Fei Li
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Xiang Liu
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Yan-Yi Xing
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou310058, China
| | - Ying Huang
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Yi-Fan Wang
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
- Precision Medicine Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117599, Singapore
| | - Tao Cheng
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Dan Wang
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Si-Han Chen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310058, China
- Liangzhu Laboratory, Ministry of Education Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou311121, China
| | - Yi-Jian Chen
- Institute of Cell and Developmental Biology, Zhejiang University School of Life Sciences, Hangzhou310058, China
| | - Guan-Nan Jiang
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Fu-I Lu
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - Yu Feng
- Department of Biophysics and Infectious Disease of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Xiao Huang
- Institute of Cell and Developmental Biology, Zhejiang University School of Life Sciences, Hangzhou310058, China
| | - Jun Ma
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Wei Liu
- Department of Metabolic Medicine, International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu32200, China
| | - Ge Bai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310058, China
- Liangzhu Laboratory, Ministry of Education Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou311121, China
| | - Peng-Fei Xu
- Women's Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou310058, China
| |
Collapse
|
23
|
Liu Y, Zhang Y, Chen S, Zhong X, Liu Q. Effect of LGR4/EGFR signaling on cell growth and cancer stem cell-like characteristics in liver cancer. Cytokine 2023; 165:156185. [PMID: 37001327 DOI: 10.1016/j.cyto.2023.156185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/06/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Leucine-rich repeat containing G-protein-coupled receptors 4 (LGR4) participates in tumor progression, invasion, and metastasis. Our study aimed to investigate the effect of LGR4 with epidermal growth factor receptor (EGFR) in HCC cells. METHODS We employed Hep3B and Huh7 cells to conduct our research. Comprehensive biological activities were characterized by CCK8 and transwell assay. Molecular biology techniques were used to determine the expression of proteins. Hep3B was employed to conduct subcutaneous tumor in mice. The tumor growth and the expression levels of proteins were assessed. RESULTS LGR4 overexpression could promote the cells proliferation, migration, and invasion ability, while siLGR4 and siEGFR could inhibit cells biological activities. In addition, LGR4 overexpression promoted the expression levels of RSPO2, β-catenin, EGFR and cancer stem cells (CSCs) markers, whereas silence of LGR4 or EGFR could diminish the expression levels of β-catenin and CSCs markers. Furthermore, knockdown of LGR4 or EGFR also inhibited tumor growth and reduced the expression levels of RSPO2, CD133, CD44, Nanog, β-catenin in vivo. CONCLUSION Our data suggest that LGR4 /EGFR signaling in HCC leads to induce tumor growth, which then contributes to stem cell characteristics. It maybe a new perspective for the targeted therapy of HCC treatment.
Collapse
Affiliation(s)
- Yanguo Liu
- Department Hepatobiliary and Pancreatic Surgery, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Yongming Zhang
- Department Hepatobiliary and Pancreatic Surgery, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Sen Chen
- Department Hepatobiliary and Pancreatic Surgery, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Xinning Zhong
- Department Hepatobiliary and Pancreatic Surgery, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Qing Liu
- Department Hepatobiliary and Pancreatic Surgery, Yantaishan Hospital, Yantai 264003, Shandong, China.
| |
Collapse
|
24
|
Sun Y, Zhang J, Hong J, Zhang Z, Lu P, Gao A, Ni M, Zhang Z, Yang H, Shen J, Lu J, Xue W, Lv Q, Bi Y, Zeng YA, Gu W, Ning G, Wang W, Liu R, Wang J. Human RSPO1 Mutation Represses Beige Adipocyte Thermogenesis and Contributes to Diet-Induced Adiposity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207152. [PMID: 36755192 PMCID: PMC10131814 DOI: 10.1002/advs.202207152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/15/2023] [Indexed: 06/18/2023]
Abstract
Recent genetic evidence has linked WNT downstream mutations to fat distribution. However, the roles of WNTs in human obesity remain unclear. Here, the authors screen all Wnt-related paracrine factors in 1994 obese cases and 2161 controls using whole-exome sequencing (WES) and identify that 12 obese patients harbor the same mutations in RSPO1 (p.R219W/Q) predisposing to human obesity. RSPO1 is predominantly expressed in visceral fat, primarily in the fibroblast cluster, and is increased with adiposity. Mice overexpressing human RSPO1 in adipose tissues develop obesity under a high-fat diet (HFD) due to reduced brown/beige fat thermogenesis. In contrast, Rspo1 ablation resists HFD-induced adiposity by increasing thermogenesis. Mechanistically, RSPO1 overexpression or administration significantly inhibits adipocyte mitochondrial respiration and thermogenesis via LGR4-Wnt/β-catenin signaling pathway. Importantly, humanized knockin mice carrying the hotspot mutation (p.R219W) display suppressed thermogenesis and recapitulate the adiposity feature of obese carriers. The mutation disrupts RSPO1's electrostatic interaction with the extracellular matrix, leading to excessive RSPO1 release that activates LGR4-Wnt/β-catenin signaling and attenuates thermogenic capacity in differentiated beige adipocytes. Therefore, these findings identify that gain-of-function mutations and excessive expression of RSPO1, acting as a paracrine Wnt activator, suppress fat thermogenesis and contribute to obesity in humans.
Collapse
Affiliation(s)
- Yingkai Sun
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Juan Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Jie Hong
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Zhongyun Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Peng Lu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Aibo Gao
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Mengshan Ni
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Zhiyin Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Huanjie Yang
- BGI GenomicsBGI‐ShenzhenShenzhen860755P. R. China
| | - Juan Shen
- BGI GenomicsBGI‐ShenzhenShenzhen860755P. R. China
| | - Jieli Lu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Wenzhi Xue
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Qianqian Lv
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Yufang Bi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Yi Arial Zeng
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell ScienceInstitute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031P. R. China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Guang Ning
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Weiqing Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Ruixin Liu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| |
Collapse
|
25
|
Chopra A, Song J, Weiner J, Keceli HG, Dincer PR, Cruz R, Carracedo A, Blanco J, Dommisch H, Schaefer AS. RSPO4 is a potential risk gene of stages III-IV, grade C periodontitis through effects on innate immune response and oral barrier integrity. J Clin Periodontol 2023; 50:476-486. [PMID: 36507580 DOI: 10.1111/jcpe.13758] [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: 06/03/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022]
Abstract
AIM R-spondin 4 (RSPO4) is a suggestive risk gene of stage III-IV, grade C periodontitis and upregulated in gingiva of mice resistant to bacteria-induced alveolar bone loss. We aimed to replicate the association, identify and characterize the putative causal variant(s) and molecular effects, and understand the downstream effects of RSPO4 upregulation. MATERIALS AND METHODS We performed a two-step association study for RSPO4 with imputed genotypes of a German-Dutch (896 stage III-IV, grade C periodontitis cases, 7104 controls) and Spanish sample (441 cases and 1141 controls). We analysed the allelic effects on transcription factor binding sites with reporter gene and antibody electrophoretic mobility shift assays. We used CRISPR/dCas9 activation and RNA sequencing to pinpoint RSPO4 as the target gene and to analyse downstream effects. RESULTS RSPO4 was associated with periodontitis (rs6056178, pmeta = 4.6 × 10-5 ). rs6056178 contains a GATA-binding motif. The rs6056178 T-allele abolished reporter activity (p = .004) and reduced GATA binding (-14.5%). CRISPRa of the associated region increased RSPO4 expression (25.8 ± 6.5-fold, p = .003). RSPO4 activation showed strongest induction of Gliomedin (439-fold) and Mucin 21 (178-fold) and of the gene set "response to interferon-alpha" (area under the curve [AUC] = 0.8, p < 5 × 10-6 ). The most repressed gene set was "extracellular matrix interactions" (AUC = 0.8, padj = .00016). CONCLUSION RSPO4 is a potential periodontitis risk gene and modifies host defence and barrier integrity.
Collapse
Affiliation(s)
- Avneesh Chopra
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Jiahui Song
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - January Weiner
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Huseyin G Keceli
- Department of Periodontology, Faculty of Dentistry, Hacettepe University, Ankara, Turkey
| | - Pervin R Dincer
- Faculty of Medicine, Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Raquel Cruz
- Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- CIBERER-Instituto de Salud Carlos III, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Angel Carracedo
- Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- CIBERER-Instituto de Salud Carlos III, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan Blanco
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Henrik Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Arne S Schaefer
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
26
|
Boer LL, Kircher SG, Rehder H, Behunova J, Winter E, Ringl H, Scharrer A, de Boer E, Oostra RJ. History and highlights of the teratological collection in the Narrenturm, Vienna (Austria). Am J Med Genet A 2023; 191:1301-1324. [PMID: 36806455 DOI: 10.1002/ajmg.a.63153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023]
Abstract
The collection of the Narrenturm in Vienna houses and maintains more than 50,000 objects including approximately 1200 teratological specimens; making it one of the biggest collections of specimens from human origin in Europe. The existence of this magnificent collection-representing an important resource for dysmorphology research, mostly awaiting contemporary diagnoses-is not widely known in the scientific community. Here, we show that the Narrenturm harbors a wealth of specimens with (exceptionally) rare congenital anomalies. These museums can be seen as physical repositories of human malformation, covering hundreds of years of dedicated collecting and preserving, thereby creating unique settings that can be used to expand our knowledge of developmental conditions that have to be preserved for future generations of scientists.
Collapse
Affiliation(s)
- Lucas L Boer
- Department of Imaging, Section Anatomy and Museum for Anatomy and Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Susanne Gerit Kircher
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Helga Rehder
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Jana Behunova
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Eduard Winter
- Pathologisch-Anatomische Sammlung im Narrenturm-NHM, Vienna, Austria
| | - Helmut Ringl
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Anke Scharrer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Elke de Boer
- Department of Human Genetics, Radboudumc, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Roelof-Jan Oostra
- Department of Medical Biology, Section Clinical Anatomy and Embryology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
27
|
Craig SEL, Michalski MN, Williams BO. Got WNTS? Insight into bone health from a WNT perspective. Curr Top Dev Biol 2023; 153:327-346. [PMID: 36967199 DOI: 10.1016/bs.ctdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
WNT signaling, essential for many aspects of development, is among the most commonly altered pathways associated with human disease. While initially studied in cancer, dysregulation of WNT signaling has been determined to be essential for skeletal development and the maintenance of bone health throughout life. In this review, we discuss the role of Wnt signaling in bone development and disease with a particular focus on two areas. First, we discuss the roles of WNT signaling pathways in skeletal development, with an emphasis on congenital and idiopathic skeletal syndromes and diseases that are associated with genetic variations in WNT signaling components. Next, we cover a topic that has long been an interest of our laboratory, how high and low levels of WNT signaling affects the establishment and maintenance of healthy bone mass. We conclude with a discussion of the status of WNT-based therapeutics in the treatment of skeletal disease.
Collapse
Affiliation(s)
- Sonya E L Craig
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Megan N Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Bart O Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW To provide an update on the current understanding of the role of wingless/integrase-1 (Wnt) signaling in pediatric allergic asthma and other pediatric lung diseases. RECENT FINDINGS The Wnt signaling pathway is critical for normal lung development. Genetic and epigenetic human studies indicate a link between Wnt signaling and the development and severity of asthma in children. Mechanistic studies using animal models of allergic asthma demonstrate a key role for Wnt signaling in allergic airway inflammation and remodeling. More recently, data on bronchopulmonary dysplasia (BPD) pathogenesis points to the Wnt signaling pathway as an important regulator. SUMMARY Current data indicates that the Wnt signaling pathway is an important mediator in allergic asthma and BPD pathogenesis. Further studies are needed to characterize the roles of individual Wnt signals in childhood disease, and to identify potential novel therapeutic targets to slow or prevent disease processes.
Collapse
Affiliation(s)
- Nooralam Rai
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Jeanine D’Armiento
- Department of Anesthesiology, Medicine, and Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
29
|
Tovar A, Smith GJ, Nalesnik MB, Thomas JM, McFadden KM, Harkema JR, Kelada SNP. A Locus on Chromosome 15 Contributes to Acute Ozone-induced Lung Injury in Collaborative Cross Mice. Am J Respir Cell Mol Biol 2022; 67:528-538. [PMID: 35816602 PMCID: PMC9651200 DOI: 10.1165/rcmb.2021-0326oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Ozone (O3)-induced respiratory toxicity varies considerably within the human population and across inbred mouse strains, indicative of gene-environment interactions (GxE). Though previous studies have identified several quantitative trait loci (QTL) and candidate genes underlying responses to O3 exposure, precise mechanisms of susceptibility remain incompletely described. We sought to update our understanding of the genetic architecture of O3 responsiveness using the Collaborative Cross (CC) recombinant inbred mouse panel. We evaluated hallmark O3-induced inflammation and injury phenotypes in 56 CC strains after exposure to filtered air or 2 ppm O3, and performed focused genetic analysis of variation in lung injury, as reflected by protein in lung lavage fluid. Strain-dependent responses to O3 were clear, and QTL mapping revealed two novel loci on Chr (Chromosomes) 10 (peak, 26.2 Mb; 80% confidence interval [CI], 24.6-43.6 Mb) and 15 (peak, 47.1 Mb; 80% CI, 40.2-54.9 Mb), the latter surpassing the 95% significance threshold. At the Chr 15 locus, C57BL/6J and CAST/EiJ founder haplotypes were associated with higher lung injury responses compared with all other CC founder haplotypes. With further statistical analysis and a weight of evidence approach, we delimited the Chr 15 QTL to an ∼2 Mb region containing 21 genes (10 protein coding) and nominated three candidate genes, namely Oxr1, Rspo2, and Angpt1. Gene and protein expression data further supported Oxr1 and Angpt1 as priority candidate genes. In summary, we have shown that O3-induced lung injury is modulated by genetic variation, identified two high priority candidate genes, and demonstrated the value of the CC for detecting GxE.
Collapse
Affiliation(s)
- Adelaide Tovar
- Department of Genetics
- Curriculum in Genetics and Molecular Biology, and
| | - Gregory J. Smith
- Department of Genetics
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Morgan B. Nalesnik
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | | | | | - Jack R. Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Samir N. P. Kelada
- Department of Genetics
- Curriculum in Genetics and Molecular Biology, and
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
30
|
Olbertová K, Hrčkulák D, Kříž V, Jesionek W, Kubovčiak J, Ešner M, Kořínek V, Buchtová M. Role of LGR5-positive mesenchymal cells in craniofacial development. Front Cell Dev Biol 2022; 10:810527. [PMID: 36133922 PMCID: PMC9484000 DOI: 10.3389/fcell.2022.810527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 08/03/2022] [Indexed: 11/28/2022] Open
Abstract
Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5), a Wnt pathway member, has been previously recognised as a stem cell marker in numerous epithelial tissues. In this study, we used Lgr5-EGFP-CreERT2 mice to analyse the distribution of LGR5-positive cells during craniofacial development. LGR5 expressing cells were primarily located in the mesenchyme adjacent to the craniofacial epithelial structures undergoing folding, such as the nasopharyngeal duct, lingual groove, and vomeronasal organ. To follow the fate of LGR5-positive cells, we performed lineage tracing using an inducible Cre knock-in allele in combination with Rosa26-tdTomato reporter mice. The slight expansion of LGR5-positive cells was found around the vomeronasal organ, in the nasal cavity, and around the epithelium in the lingual groove. However, most LGR5 expressing cells remained in their original location, possibly supporting their signalling function for adjacent epithelium rather than exerting their role as progenitor cells for the craniofacial structures. Moreover, Lgr5 knockout mice displayed distinct defects in LGR5-positive areas, especially in the reduction of the nasopharyngeal duct, the alteration of the palatal shelves shape, abnormal epithelial folding in the lingual groove area, and the disruption of salivary gland development. The latter defect manifested as an atypical number and localisation of the glandular ducts. The gene expression of several Wnt pathway members (Rspo1-3, Axin2) was altered in Lgr5-deficient animals. However, the difference was not found in sorted EGFP-positive cells obtained from Lgr5 +/+ and Lgr5 -/- animals. Expression profiling of LGR5-positive cells revealed the expression of several markers of mesenchymal cells, antagonists, as well as agonists, of Wnt signalling, and molecules associated with the basal membrane. Therefore, LGR5-positive cells in the craniofacial area represent a very specific population of mesenchymal cells adjacent to the epithelium undergoing folding or groove formation. Our results indicate a possible novel role of LGR5 in the regulation of morphogenetic processes during the formation of complex epithelial structures in the craniofacial areas, a role which is not related to the stem cell properties of LGR5-positive cells as was previously defined for various epithelial tissues.
Collapse
Affiliation(s)
- Kristýna Olbertová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Dušan Hrčkulák
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Vítězslav Kříž
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Wojciech Jesionek
- Cellular Imaging Core Facility, Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Milan Ešner
- Cellular Imaging Core Facility, Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
| | - Vladimír Kořínek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
31
|
Yang L, Yue W, Zhang H, Gao Y, Yang L, Li L. The role of roof plate-specific spondins in liver homeostasis and disease. LIVER RESEARCH 2022; 6:139-145. [PMID: 39958194 PMCID: PMC11791806 DOI: 10.1016/j.livres.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 02/16/2023]
Abstract
As evolutionarily conserved signals, roof plate-specific spondins (R-spondins; RSPOs) are a family with four members (RSPO1-4) exerting distinctly different functions. RSPOs have five receptors and correlate with different signaling pathways through these receptors and then perform various functions. Moreover, their best-known molecular function is the capacity to enhance WNT signaling pathways, which play critical roles in several processes. A recent study shows that RSPOs not only potentiate the WNT/beta (β)-catenin signaling pathway but are also involved in the WNT/planar cell polarity signaling pathway. RSPOs influence liver homeostasis and the development of multiple liver diseases. RSPO1 increases cell proliferation, protects hepatocytes from injury, improves liver regenerative potential, and affects liver metabolic zonation. RSPO2 not only regulates proliferation-associated genes and promotes differentiation in the liver but also participates in liver fibrosis through the WNT/β-catenin signaling pathway. RSPO3 is a key determinant of proper liver function, such as promoting hepatocyte regeneration and maintaining liver zonation. RSPO3 is upregulated in liver fibrosis and livers of patients with non-alcoholic steatohepatitis. Besides, RSPO2 and RSPO3 are confirmed as oncogenes and involved in the occurrence of liver cancer. The role of RSPO4 in the liver remains unclear. In this review, the structural and biochemical properties of RSPOs and their receptors and their roles in liver homeostasis and disease are summarized.
Collapse
Affiliation(s)
- Le Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Wenhui Yue
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Hang Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Yue Gao
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Tsotakos N, Ahmed I, Umstead TM, Imamura Y, Yau E, Silveyra P, Chroneos ZC. All trans-retinoic acid modulates hyperoxia-induced suppression of NF-kB-dependent Wnt signaling in alveolar A549 epithelial cells. PLoS One 2022; 17:e0272769. [PMID: 35947545 PMCID: PMC9365139 DOI: 10.1371/journal.pone.0272769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Despite recent advances in perinatal medicine, bronchopulmonary dysplasia (BPD) remains the most common complication of preterm birth. Inflammation, the main cause for BPD, results in arrested alveolarization. All trans-retinoic acid (ATRA), the active metabolite of Vitamin A, facilitates recovery from hyperoxia induced cell damage. The mechanisms involved in this response, and the genes activated, however, are poorly understood. In this study, we investigated the mechanisms of action of ATRA in human lung epithelial cells exposed to hyperoxia. We hypothesized that ATRA reduces hyperoxia-induced inflammatory responses in A549 alveolar epithelial cells. METHODS A549 cells were exposed to hyperoxia with or without treatment with ATRA, followed by RNA-seq analysis. RESULTS Transcriptomic analysis of A549 cells revealed ~2,000 differentially expressed genes with a higher than 2-fold change. Treatment of cells with ATRA alleviated some of the hyperoxia-induced changes, including Wnt signaling, cell adhesion and cytochrome P450 genes, partially through NF-κB signaling. DISCUSSION/CONCLUSION Our findings support the idea that ATRA supplementation may decrease hyperoxia-induced disruption of the neonatal respiratory epithelium and alleviate development of BPD.
Collapse
Affiliation(s)
- Nikolaos Tsotakos
- School of Science, Engineering, and Technology, Penn State Harrisburg, Middletown, Pennsylvania, United States
| | - Imtiaz Ahmed
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Todd M. Umstead
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Yuka Imamura
- Departments of Pharmacology and Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- Institute of Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Eric Yau
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Patricia Silveyra
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States of America
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Zissis C. Chroneos
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- Institute of Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
33
|
Zhang Z, Chai R. Hear the sounds: The role of G Protein-Coupled Receptors in the cochlea. Am J Physiol Cell Physiol 2022; 323:C1088-C1099. [PMID: 35938679 DOI: 10.1152/ajpcell.00453.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sound is converted by hair cells in the cochlea into electrical signals, which are transmitted by spiral ganglion neurons (SGNs) and heard by the auditory cortex. G protein-coupled receptors (GPCRs) are crucial receptors that regulate a wide range of physiological functions in different organ and tissues. The research of GPCRs in the cochlea is essential for the understanding of the cochlea development, hearing disorders, and the treatment for hearing loss. Recently, several GPCRs have been found to play important roles in the cochlea. Frizzleds and Lgrs are dominant GPCRs that regulate stem cell self-renew abilities. Moreover, Frizzleds and Celsrs have been demonstrated to play core roles in the modulation of cochlear planar cell polarity (PCP). In addition, hearing loss can be caused by mutations of certain GPCRs, such as Vlgr1, Gpr156, S1P2 and Gpr126. And A1, A2A and CB2 activation by agonists have protective functions on noise- or drug-induced hearing loss. Here, we review the key findings of GPCR in the cochlea, and discuss the role of GPCR in the cochlea, such as stem cell fate, PCP, hearing loss, and hearing protection.
Collapse
Affiliation(s)
- Zhong Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| |
Collapse
|
34
|
Hein RFC, Wu JH, Holloway EM, Frum T, Conchola AS, Tsai YH, Wu A, Fine AS, Miller AJ, Szenker-Ravi E, Yan KS, Kuo CJ, Glass I, Reversade B, Spence JR. R-SPONDIN2 + mesenchymal cells form the bud tip progenitor niche during human lung development. Dev Cell 2022; 57:1598-1614.e8. [PMID: 35679862 PMCID: PMC9283295 DOI: 10.1016/j.devcel.2022.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 01/23/2023]
Abstract
The human respiratory epithelium is derived from a progenitor cell in the distal buds of the developing lung. These "bud tip progenitors" are regulated by reciprocal signaling with surrounding mesenchyme; however, mesenchymal heterogeneity and function in the developing human lung are poorly understood. We interrogated single-cell RNA sequencing data from multiple human lung specimens and identified a mesenchymal cell population present during development that is highly enriched for expression of the WNT agonist RSPO2, and we found that the adjacent bud tip progenitors are enriched for the RSPO2 receptor LGR5. Functional experiments using organoid models, explant cultures, and FACS-isolated RSPO2+ mesenchyme show that RSPO2 is a critical niche cue that potentiates WNT signaling in bud tip progenitors to support their maintenance and multipotency.
Collapse
Affiliation(s)
- Renee F C Hein
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joshua H Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tristan Frum
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ansley S Conchola
- Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexis S Fine
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alyssa J Miller
- Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emmanuelle Szenker-Ravi
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore, A(∗)STAR, Singapore 138648, Singapore
| | - Kelley S Yan
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Departments of Medicine and Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ian Glass
- Department of Pediatrics, Genetic Medicine, University of Washington, Seattle, WA 98195, USA
| | - Bruno Reversade
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore, A(∗)STAR, Singapore 138648, Singapore; Laboratory of Human Genetics & Therapeutics, Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore; Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul, Turkey
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA.
| |
Collapse
|
35
|
Glotzer GL, Tardivo P, Tanaka EM. Canonical Wnt signaling and the regulation of divergent mesenchymal Fgf8 expression in axolotl limb development and regeneration. eLife 2022; 11:e79762. [PMID: 35587651 PMCID: PMC9154742 DOI: 10.7554/elife.79762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/06/2022] [Indexed: 11/30/2022] Open
Abstract
The expression of fibroblast growth factors (Fgf) ligands in a specialized epithelial compartment, the Apical Ectodermal Ridge (AER), is a conserved feature of limb development across vertebrate species. In vertebrates, Fgf 4, 8, 9, and 17 are all expressed in the AER. An exception to this paradigm is the salamander (axolotl) developing and regenerating limb, where key Fgf ligands are expressed in the mesenchyme. The mesenchymal expression of Amex.Fgf8 in axolotl has been suggested to be critical for regeneration. To date, there is little knowledge regarding what controls Amex.Fgf8 expression in the axolotl limb mesenchyme. A large body of mouse and chick studies have defined a set of transcription factors and canonical Wnt signaling as the main regulators of epidermal Fgf8 expression in these organisms. In this study, we address the hypothesis that alterations to one or more of these components during evolution has resulted in mesenchymal Amex.Fgf8 expression in the axolotl. To sensitively quantify gene expression with spatial precision, we combined optical clearing of whole-mount axolotl limb tissue with single molecule fluorescent in situ hybridization and a semiautomated quantification pipeline. Several candidate upstream components were found expressed in the axolotl ectoderm, indicating that they are not direct regulators of Amex.Fgf8 expression. We found that Amex.Wnt3a is expressed in axolotl limb epidermis, similar to chicken and mouse. However, unlike in amniotes, Wnt target genes are activated preferentially in limb mesenchyme rather than in epidermis. Inhibition and activation of Wnt signaling results in downregulation and upregulation of mesenchymal Amex.Fgf8 expression, respectively. These results implicate a shift in tissue responsiveness to canonical Wnt signaling from epidermis to mesenchyme as one step contributing to the unique mesenchymal Amex.Fgf8 expression seen in the axolotl.
Collapse
Affiliation(s)
- Giacomo L Glotzer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus- Vienna-Biocenter 1ViennaAustria
| | - Pietro Tardivo
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus- Vienna-Biocenter 1ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of ViennaViennaAustria
| | - Elly M Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus- Vienna-Biocenter 1ViennaAustria
| |
Collapse
|
36
|
Raslan AA, Oh YJ, Jin YR, Yoon JK. R-Spondin2, a Positive Canonical WNT Signaling Regulator, Controls the Expansion and Differentiation of Distal Lung Epithelial Stem/Progenitor Cells in Mice. Int J Mol Sci 2022; 23:ijms23063089. [PMID: 35328508 PMCID: PMC8954098 DOI: 10.3390/ijms23063089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
The lungs have a remarkable ability to regenerate damaged tissues caused by acute injury. Many lung diseases, especially chronic lung diseases, are associated with a reduced or disrupted regeneration potential of the lungs. Therefore, understanding the underlying mechanisms of the regenerative capacity of the lungs offers the potential to identify novel therapeutic targets for these diseases. R-spondin2, a co-activator of WNT/β-catenin signaling, plays an important role in embryonic murine lung development. However, the role of Rspo2 in adult lung homeostasis and regeneration remains unknown. The aim of this study is to determine Rspo2 function in distal lung stem/progenitor cells and adult lung regeneration. In this study, we found that robust Rspo2 expression was detected in different epithelial cells, including airway club cells and alveolar type 2 (AT2) cells in the adult lungs. However, Rspo2 expression significantly decreased during the first week after naphthalene-induced airway injury and was restored by day 14 post-injury. In ex vivo 3D organoid culture, recombinant RSPO2 promoted the colony formation and differentiation of both club and AT2 cells through the activation of canonical WNT signaling. In contrast, Rspo2 ablation in club and AT2 cells significantly disrupted their expansion capacity in the ex vivo 3D organoid culture. Furthermore, mice lacking Rspo2 showed significant defects in airway regeneration after naphthalene-induced injury. Our results strongly suggest that RSPO2 plays a key role in the adult lung epithelial stem/progenitor cells during homeostasis and regeneration, and therefore, it may be a potential therapeutic target for chronic lung diseases with reduced regenerative capability.
Collapse
Affiliation(s)
- Ahmed A. Raslan
- Department of Integrated Biomedical Science, Graduate School, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
| | - Youn Jeong Oh
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
| | - Yong Ri Jin
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Jeong Kyo Yoon
- Department of Integrated Biomedical Science, Graduate School, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Correspondence:
| |
Collapse
|
37
|
Baldwin AT, Kim JH, Seo H, Wallingford JB. Global analysis of cell behavior and protein dynamics reveals region-specific roles for Shroom3 and N-cadherin during neural tube closure. eLife 2022; 11:e66704. [PMID: 35244026 PMCID: PMC9010020 DOI: 10.7554/elife.66704] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
Failures of neural tube closure are common and serious birth defects, yet we have a poor understanding of the interaction of genetics and cell biology during neural tube closure. Additionally, mutations that cause neural tube defects (NTDs) tend to affect anterior or posterior regions of the neural tube but rarely both, indicating a regional specificity to NTD genetics. To better understand the regional specificity of cell behaviors during neural tube closure, we analyzed the dynamic localization of actin and N-cadherin via high-resolution tissue-level time-lapse microscopy during Xenopus neural tube closure. To investigate the regionality of gene function, we generated mosaic mutations in shroom3, a key regulator or neural tube closure. This new analytical approach elucidates several differences between cell behaviors during cranial/anterior and spinal/posterior neural tube closure, provides mechanistic insight into the function of shroom3, and demonstrates the ability of tissue-level imaging and analysis to generate cell biological mechanistic insights into neural tube closure.
Collapse
Affiliation(s)
- Austin T Baldwin
- Department of Molecular Biosciences, University of Texas at AustinAustinUnited States
| | - Juliana H Kim
- Department of Molecular Biosciences, University of Texas at AustinAustinUnited States
| | - Hyemin Seo
- Department of Molecular Biosciences, University of Texas at AustinAustinUnited States
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at AustinAustinUnited States
| |
Collapse
|
38
|
Nilsson KH, Wu J, Gustafsson KL, El Shahawy M, Koskela A, Tuukkanen J, Tuckermann J, Henning P, Lerner UH, Ohlsson C, Movérare-Skrtic S. Estradiol and RSPO3 regulate vertebral trabecular bone mass independent of each other. Am J Physiol Endocrinol Metab 2022; 322:E211-E218. [PMID: 35068191 PMCID: PMC8896994 DOI: 10.1152/ajpendo.00383.2021] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Osteoporosis is an age-dependent serious skeletal disease that leads to great suffering for the patient and high social costs, especially as the global population reaches higher age. Decreasing estrogen levels after menopause result in a substantial bone loss and increased fracture risk, whereas estrogen treatment improves bone mass in women. RSPO3, a secreted protein that modulates WNT signaling, increases trabecular bone mass and strength in the vertebrae of mice, and is associated with trabecular density and risk of distal forearm fractures in humans. The aim of the present study was to determine if RSPO3 is involved in the bone-sparing effect of estrogens. We first observed that estradiol (E2) treatment increases RSPO3 expression in bone of ovariectomized (OVX) mice, supporting a possible role of RSPO3 in the bone-sparing effect of estrogens. As RSPO3 is mainly expressed by osteoblasts in the bone, we used a mouse model devoid of osteoblast-derived RSPO3 (Runx2-creRspo3flox/flox mice) to determine if RSPO3 is required for the bone-sparing effect of E2 in OVX mice. We confirmed that osteoblast-specific RSPO3 inactivation results in a substantial reduction in trabecular bone mass and strength in the vertebrae. However, E2 increased vertebral trabecular bone mass and strength similarly in mice devoid of osteoblast-derived RSPO3 and control mice. Unexpectedly, osteoblast-derived RSPO3 was needed for the full estrogenic response on cortical bone thickness. In conclusion, although osteoblast-derived RSPO3 is a crucial regulator of vertebral trabecular bone, it is required for a full estrogenic effect on cortical, but not trabecular, bone in OVX mice. Thus, estradiol and RSPO3 regulate vertebral trabecular bone mass independent of each other.NEW & NOTEWORTHY Osteoblast-derived RSPO3 is known to be a crucial regulator of vertebral trabecular bone. Our new findings show that RSPO3 and estrogen regulate trabecular bone independent of each other, but that RSPO3 is necessary for a complete estrogenic effect on cortical bone.
Collapse
Affiliation(s)
- Karin H Nilsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin L Gustafsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Maha El Shahawy
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
39
|
Abstract
The Wnt pathway is central to a host of developmental and disease-related processes. The remarkable conservation of this intercellular signaling cascade throughout metazoan lineages indicates that it coevolved with multicellularity to regulate the generation and spatial arrangement of distinct cell types. By regulating cell fate specification, mitotic activity, and cell polarity, Wnt signaling orchestrates development and tissue homeostasis, and its dysregulation is implicated in developmental defects, cancer, and degenerative disorders. We review advances in our understanding of this key pathway, from Wnt protein production and secretion to relay of the signal in the cytoplasm of the receiving cell. We discuss the evolutionary history of this pathway as well as endogenous and synthetic modulators of its activity. Finally, we highlight remaining gaps in our knowledge of Wnt signal transduction and avenues for future research. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ellen Youngsoo Rim
- Howard Hughes Medical Institute, Department of Developmental Biology, and Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA;
| | - Hans Clevers
- Hubrecht Institute and Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, The Netherlands
| | - Roel Nusse
- Howard Hughes Medical Institute, Department of Developmental Biology, and Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA;
| |
Collapse
|
40
|
Yang L, Wang J, Gong X, Fan Q, Yang X, Cui Y, Gao X, Li L, Sun X, Li Y, Wang Y. Emerging Roles for LGR4 in Organ Development, Energy Metabolism and Carcinogenesis. Front Genet 2022; 12:728827. [PMID: 35140734 PMCID: PMC8819683 DOI: 10.3389/fgene.2021.728827] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/30/2021] [Indexed: 11/26/2022] Open
Abstract
The leucine-rich repeats containing G protein-coupled receptor 4 (LGR4) belonging to G protein-coupled receptors (GPCRs) family, had various regulatory roles at multiple cellular types and numerous targeting sites, and aberrant LGR4 signaling played crucial roles in diseases and carcinogenesis. On the basis of these facts, LGR4 may become an appealing therapeutic target for the treatment of diseases and tumors. However, a comprehensive investigation of its functions and applications was still lacking. Hence, this paper provided an overview of the molecular characteristics and signaling mechanisms of LGR4, its involvement in multiple organ development and participation in the modulation of immunology related diseases, metabolic diseases, and oxidative stress damage along with cancer progression. Given that GPCRs accounted for almost a third of current clinical drug targets, the in-depth understanding of the sophisticated connections of LGR4 and its ligands would not only enrich their regulatory networks, but also shed new light on designing novel molecular targeted drugs and small molecule blockers for revolutionizing the treatment of various diseases and tumors.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Jing Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Qiong Fan
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoming Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yunxia Cui
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoyan Gao
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Lijuan Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiao Sun
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yuhong Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| | - Yudong Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| |
Collapse
|
41
|
Lee H, Sun R, Niehrs C. Uncoupling the BMP receptor antagonist function from the WNT agonist function of R-spondin 2 using the inhibitory peptide dendrimer RW d. J Biol Chem 2022; 298:101586. [PMID: 35032551 PMCID: PMC8842081 DOI: 10.1016/j.jbc.2022.101586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/28/2022] Open
Abstract
Signaling by bone morphogenetic proteins (BMPs) plays pivotal roles in embryogenesis, adult tissue homeostasis, and disease. Recent studies revealed that the well-established WNT agonist R-spondin 2 (RSPO2) is also a BMP receptor (BMP receptor type 1A) antagonist, with roles in early Xenopus embryogenesis and human acute myeloid leukemia (AML). To uncouple the BMP antagonist function from the WNT agonist function and to promote development of AML therapeutics, here we identified a 10-mer peptide (RW) derived from the thrombospondin 1 domain of RSPO2, which specifically prevents binding between RSPO2 and BMP receptor type 1A without altering WNT signaling. We also show that a corresponding RW dendrimer (RWd) exhibiting improved half-life relieves inhibition of BMP receptor signaling by RSPO2 in human AML cells, reduces cell growth, and induces differentiation. Moreover, microinjection of RWd in Xenopus embryos ventralizes the dorsoventral embryonic patterning by upregulating BMP signaling without affecting WNT signaling. Our study corroborates the function of RSPO2 as a BMP receptor antagonist and provides a proof of concept for pharmacologically uncoupling BMP antagonist from WNT agonist functions of RSPO2 using the inhibitor peptide RWd with enhanced target selectivity and limited side effects.
Collapse
Affiliation(s)
- Hyeyoon Lee
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Rui Sun
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany; Institute of Molecular Biology (IMB), Mainz, Germany.
| |
Collapse
|
42
|
Li J, Glover JD, Zhang H, Peng M, Tan J, Mallick CB, Hou D, Yang Y, Wu S, Liu Y, Peng Q, Zheng SC, Crosse EI, Medvinsky A, Anderson RA, Brown H, Yuan Z, Zhou S, Xu Y, Kemp JP, Ho YYW, Loesch DZ, Wang L, Li Y, Tang S, Wu X, Walters RG, Lin K, Meng R, Lv J, Chernus JM, Neiswanger K, Feingold E, Evans DM, Medland SE, Martin NG, Weinberg SM, Marazita ML, Chen G, Chen Z, Zhou Y, Cheeseman M, Wang L, Jin L, Headon DJ, Wang S. Limb development genes underlie variation in human fingerprint patterns. Cell 2022; 185:95-112.e18. [PMID: 34995520 PMCID: PMC8740935 DOI: 10.1016/j.cell.2021.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Fingerprints are of long-standing practical and cultural interest, but little is known about the mechanisms that underlie their variation. Using genome-wide scans in Han Chinese cohorts, we identified 18 loci associated with fingerprint type across the digits, including a genetic basis for the long-recognized "pattern-block" correlations among the middle three digits. In particular, we identified a variant near EVI1 that alters regulatory activity and established a role for EVI1 in dermatoglyph patterning in mice. Dynamic EVI1 expression during human development supports its role in shaping the limbs and digits, rather than influencing skin patterning directly. Trans-ethnic meta-analysis identified 43 fingerprint-associated loci, with nearby genes being strongly enriched for general limb development pathways. We also found that fingerprint patterns were genetically correlated with hand proportions. Taken together, these findings support the key role of limb development genes in influencing the outcome of fingerprint patterning.
Collapse
Affiliation(s)
- Jinxi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Haiguo Zhang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Meifang Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Jingze Tan
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Chandana Basu Mallick
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK; Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Dan Hou
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yajun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Sijie Wu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yu Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Shijie C Zheng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Edie I Crosse
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Helen Brown
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, PRC
| | - Shen Zhou
- Shanghai Foreign Language School, Shanghai 200083, PRC
| | - Yanqing Xu
- Forest Ridge School of the Sacred Heart, Bellevue, WA 98006, USA
| | - John P Kemp
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | - Yvonne Y W Ho
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | - Danuta Z Loesch
- Psychology Department, La Trobe University, Melbourne, VIC, Australia
| | | | | | | | - Xiaoli Wu
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ruogu Meng
- Center for Data Science in Health and Medicine, Peking University, Beijing 100191, PRC
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, PRC
| | - Jonathan M Chernus
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Katherine Neiswanger
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David M Evans
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Sarah E Medland
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | | | - Seth M Weinberg
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Anthropology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mary L Marazita
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gang Chen
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Yong Zhou
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PRC
| | - Michael Cheeseman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Lan Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai 200438, PRC.
| | - Denis J Headon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, PRC.
| |
Collapse
|
43
|
Lebensohn AM, Bazan JF, Rohatgi R. Receptor control by membrane-tethered ubiquitin ligases in development and tissue homeostasis. Curr Top Dev Biol 2022; 150:25-89. [PMID: 35817504 DOI: 10.1016/bs.ctdb.2022.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Paracrine cell-cell communication is central to all developmental processes, ranging from cell diversification to patterning and morphogenesis. Precise calibration of signaling strength is essential for the fidelity of tissue formation during embryogenesis and tissue maintenance in adults. Membrane-tethered ubiquitin ligases can control the sensitivity of target cells to secreted ligands by regulating the abundance of signaling receptors at the cell surface. We discuss two examples of this emerging concept in signaling: (1) the transmembrane ubiquitin ligases ZNRF3 and RNF43 that regulate WNT and bone morphogenetic protein receptor abundance in response to R-spondin ligands and (2) the membrane-recruited ubiquitin ligase MGRN1 that controls Hedgehog and melanocortin receptor abundance. We focus on the mechanistic logic of these systems, illustrated by structural and protein interaction models enabled by AlphaFold. We suggest that membrane-tethered ubiquitin ligases play a widespread role in remodeling the cell surface proteome to control responses to extracellular ligands in diverse biological processes.
Collapse
|
44
|
Nagano K, Yamana K, Saito H, Kiviranta R, Pedroni AC, Raval D, Niehrs C, Gori F, Baron R. R-spondin 3 deletion induces Erk phosphorylation to enhance Wnt signaling and promote bone formation in the appendicular skeleton. eLife 2022; 11:84171. [PMID: 36321691 PMCID: PMC9681208 DOI: 10.7554/elife.84171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
Abstract
Activation of Wnt signaling leads to high bone density. The R-spondin family of four secreted glycoproteins (Rspo1-4) amplifies Wnt signaling. In humans, RSPO3 variants are strongly associated with bone density. Here, we investigated the role of Rspo3 in skeletal homeostasis in mice. Using a comprehensive set of mouse genetic and mechanistic studies, we show that in the appendicular skeleton, Rspo3 haplo-insufficiency and Rspo3 targeted deletion in Runx2+ osteoprogenitors lead to an increase in trabecular bone mass, with increased number of osteoblasts and bone formation. In contrast and highlighting the complexity of Wnt signaling in the regulation of skeletal homeostasis, we show that Rspo3 deletion in osteoprogenitors results in the opposite phenotype in the axial skeleton, i.e., low vertebral trabecular bone mass. Mechanistically, Rspo3 deficiency impairs the inhibitory effect of Dkk1 on Wnt signaling activation and bone mass. We demonstrate that Rspo3 deficiency leads to activation of Erk signaling which in turn, stabilizes β-catenin and Wnt signaling activation. Our data demonstrate that Rspo3 haplo-insufficiency/deficiency boosts canonical Wnt signaling by activating Erk signaling, to favor osteoblastogenesis, bone formation, and bone mass.
Collapse
Affiliation(s)
- Kenichi Nagano
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | - Kei Yamana
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | - Hiroaki Saito
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | - Riku Kiviranta
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | | | - Dhairya Raval
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | - Christof Niehrs
- German Cancer Research Center, DKFZ-ZMBH AllianceHeidelbergGermany,Institute of Molecular Biology (IMB)MainzGermany
| | - Francesca Gori
- School of Dental Medicine, Harvard UniversityBostonUnited States
| | - Roland Baron
- School of Dental Medicine, Harvard UniversityBostonUnited States,Department of Medicine, Harvard Medical SchoolBostonUnited States,Endocrine Unit, Massachusetts General HospitalBostonUnited States
| |
Collapse
|
45
|
Szenker-Ravi E, Ott T, Khatoo M, Moreau de Bellaing A, Goh WX, Chong YL, Beckers A, Kannesan D, Louvel G, Anujan P, Ravi V, Bonnard C, Moutton S, Schoen P, Fradin M, Colin E, Megarbane A, Daou L, Chehab G, Di Filippo S, Rooryck C, Deleuze JF, Boland A, Arribard N, Eker R, Tohari S, Ng AYJ, Rio M, Lim CT, Eisenhaber B, Eisenhaber F, Venkatesh B, Amiel J, Crollius HR, Gordon CT, Gossler A, Roy S, Attie-Bitach T, Blum M, Bouvagnet P, Reversade B. Discovery of a genetic module essential for assigning left-right asymmetry in humans and ancestral vertebrates. Nat Genet 2022; 54:62-72. [PMID: 34903892 DOI: 10.1038/s41588-021-00970-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 10/14/2021] [Indexed: 01/24/2023]
Abstract
The vertebrate left-right axis is specified during embryogenesis by a transient organ: the left-right organizer (LRO). Species including fish, amphibians, rodents and humans deploy motile cilia in the LRO to break bilateral symmetry, while reptiles, birds, even-toed mammals and cetaceans are believed to have LROs without motile cilia. We searched for genes whose loss during vertebrate evolution follows this pattern and identified five genes encoding extracellular proteins, including a putative protease with hitherto unknown functions that we named ciliated left-right organizer metallopeptide (CIROP). Here, we show that CIROP is specifically expressed in ciliated LROs. In zebrafish and Xenopus, CIROP is required solely on the left side, downstream of the leftward flow, but upstream of DAND5, the first asymmetrically expressed gene. We further ascertained 21 human patients with loss-of-function CIROP mutations presenting with recessive situs anomalies. Our findings posit the existence of an ancestral genetic module that has twice disappeared during vertebrate evolution but remains essential for distinguishing left from right in humans.
Collapse
Affiliation(s)
- Emmanuelle Szenker-Ravi
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore.
| | - Tim Ott
- Institute of Biology, University of Hohenheim, Stuttgart, Germany
| | - Muznah Khatoo
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore
| | - Anne Moreau de Bellaing
- Laboratoire de Cardiogénétique, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Wei Xuan Goh
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore
| | - Yan Ling Chong
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Anja Beckers
- Institute for Molecular Biology, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
| | - Darshini Kannesan
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore
| | - Guillaume Louvel
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
- Écologie, Systématique et Évolution, UMR 8079 CNRS - Université Paris-Saclay - AgroParisTech, Orsay, France
| | - Priyanka Anujan
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
- Institute of Reproductive and Developmental Biology, Hammersmith Hospital, Imperial College, London, UK
| | - Vydianathan Ravi
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
| | - Carine Bonnard
- Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Singapore
| | - Sébastien Moutton
- CPDPN, Pôle mère enfant, Maison de Santé Protestante Bordeaux Bagatelle, Talence, France
| | | | - Mélanie Fradin
- Service de Génétique Médicale, Hôpital Sud, CHU de Rennes, Rennes, France
| | - Estelle Colin
- Service de Génétique Médicale, CHU d'Angers, Angers, France
| | - André Megarbane
- Department of Human Genetics, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Institut Jérôme LEJEUNE, Paris, France
| | - Linda Daou
- Department of Pediatric Cardiology, Hôtel Dieu de France University Medical Center, Saint Joseph University, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| | - Ghassan Chehab
- Department of Pediatric Cardiology, Hôtel Dieu de France University Medical Center, Saint Joseph University, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
- Department of Pediatrics, Lebanese University, Faculty of Medical Sciences, Hadath, Greater Beirut, Lebanon
| | - Sylvie Di Filippo
- Service de Cardiologie Pédiatrique, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Caroline Rooryck
- Service de Génétique, University of Bordeaux, MRGM, INSERM U1211, CHU de Bordeaux, Bordeaux, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Nicolas Arribard
- Service de Cardiologie Pédiatrique, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Brussels, Belgium
| | - Rukiye Eker
- Pediatrics Department, Pediatric Cardiology Division, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Sumanty Tohari
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
| | - Alvin Yu-Jin Ng
- Molecular Diagnosis Centre (MDC), National University Hospital (NUH), Singapore, Singapore
| | - Marlène Rio
- Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Developmental Brain Disorders Laboratory, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Chun Teck Lim
- Bioinformatics Institute (BII), A*STAR, Singapore, Singapore
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), A*STAR, Singapore, Singapore
| | - Birgit Eisenhaber
- Bioinformatics Institute (BII), A*STAR, Singapore, Singapore
- Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), A*STAR, Singapore, Singapore
- Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore, Singapore
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
- Department of Pediatrics, National University of Singapore (NUS), Singapore, Singapore
| | - Jeanne Amiel
- Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Laboratory of Embryology and Genetics of Malformations, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Hugues Roest Crollius
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Christopher T Gordon
- Laboratory of Embryology and Genetics of Malformations, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Achim Gossler
- Institute for Molecular Biology, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
| | - Sudipto Roy
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore
- Department of Pediatrics, National University of Singapore (NUS), Singapore, Singapore
- Department of Biological Sciences, National University of Singapore (NUS), Singapore, Singapore
| | - Tania Attie-Bitach
- Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Laboratory of Genetics and Development of the Cerebral Cortex, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Martin Blum
- Institute of Biology, University of Hohenheim, Stuttgart, Germany.
| | | | - Bruno Reversade
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A*STAR, Singapore, Singapore.
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, Singapore.
- Department of Pediatrics, National University of Singapore (NUS), Singapore, Singapore.
- Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul, Turkey.
| |
Collapse
|
46
|
Tang Y, Xu Q, Hu L, Yan X, Feng X, Yokota A, Wang W, Zhan D, Krishnamurthy D, Ochayon DE, Wen L, Huo L, Zeng H, Luo Y, Huang LF, Wunderlich M, Zhang J, Vivier E, Zhou J, Waggoner SN, Huang G. Tumor Microenvironment-Derived R-spondins Enhance Antitumor Immunity to Suppress Tumor Growth and Sensitize for Immune Checkpoint Blockade Therapy. Cancer Discov 2021; 11:3142-3157. [PMID: 34193438 PMCID: PMC8716674 DOI: 10.1158/2159-8290.cd-20-0833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 05/07/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells and T cells are key effectors of antitumor immune responses and major targets of checkpoint inhibitors. In multiple cancer types, we find that the expression of Wnt signaling potentiator R-spondin genes (e.g., RSPO3) is associated with favorable prognosis and positively correlates with gene signatures of both NK cells and T cells. Although endothelial cells and cancer-associated fibroblasts comprise the R-spondin 3-producing cells, NK cells and T cells correspondingly express the R-spondin 3 receptor LGR6 within the tumor microenvironment (TME). Exogenous expression or intratumor injection of R-spondin 3 in tumors enhanced the infiltration and function of cytotoxic effector cells, which led to tumor regression. NK cells and CD8+ T cells independently and cooperatively contributed to R-spondin 3-induced control of distinct tumor types. The effect of R-spondin 3 was mediated in part through upregulation of MYC and ribosomal biogenesis. Importantly, R-spondin 3 expression enhanced tumor sensitivity to anti-PD-1 therapy, thereby highlighting new therapeutic avenues. SIGNIFICANCE Our study identifies novel targets in enhancing antitumor immunity and sensitizing immune checkpoint inhibition, which provides a rationale for developing new immunotherapies against cancers. It also offers mechanistic insights on Wnt signaling-mediated modulation of anticancer immunity in the TME and implications for a putative R-spondin-LGR6 axis in regulating NK-cell biology. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Yuting Tang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Qian Xu
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liang Hu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Xiaomei Yan
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Xiaomin Feng
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Asumi Yokota
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Weinan Wang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Di Zhan
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Durga Krishnamurthy
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - David E. Ochayon
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Lijun Wen
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Huo
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huimin Zeng
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Yingwan Luo
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - L. Frank Huang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Jiwang Zhang
- Oncology Institute, Loyola University Chicago, Maywood, IL 60153
- Department of Pathology, Loyola University Chicago, Maywood, IL 60153
| | - Eric Vivier
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Immunology, Marseille Immunopole, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, France
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Stephen N. Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| |
Collapse
|
47
|
Choi RB, Robling AG. The Wnt pathway: An important control mechanism in bone's response to mechanical loading. Bone 2021; 153:116087. [PMID: 34271473 PMCID: PMC8478810 DOI: 10.1016/j.bone.2021.116087] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/01/2021] [Accepted: 06/21/2021] [Indexed: 10/25/2022]
Abstract
The conversion of mechanical energy into biochemical changes within living cells is process known as mechanotransduction. Bone is a quintessential tissue for studying the molecular mechanisms of mechanotransduction, as the skeleton's mechanical competence is crucial for vertebrate movement. Bone cell mechanotransduction is facilitated by a number of cell biological pathways, one of the most prominent of which is the Wnt signaling cascade. The Wnt co-receptor Lrp5 has been identified as a crucial protein for mechanical signaling in bone, and modifiers of Lrp5 activity play important roles in mediating signaling efficiency through Lrp5, including sclerostin, Dkk1, and the co-receptor Lrp4. Mechanical regulation of sclerostin is mediated by certain members of the Hdac family. Other mechanisms that influence Wnt signaling-some of which are mechanoresponsive-are coming to light, including R-spondins and their role in organizing the Rnf43/Znrf3 and Lgr4/5/6 complex that liberates Lrp5. While the identity of the key Wnt proteins involved in bone cell mechanical signaling are elusive, the likely pool of key players is narrowing. Identification of Wnt-based molecular targets that can be modulated pharmacologically to make mechanical stimulation (e.g., exercise) more beneficial is an emerging approach to improving skeletal integrity and reducing fracture risk.
Collapse
Affiliation(s)
- Roy B Choi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
48
|
Cheng YY, Yang X, Gao X, Song SX, Yang MF, Xie FM. LGR6 promotes glioblastoma malignancy and chemoresistance by activating the Akt signaling pathway. Exp Ther Med 2021; 22:1364. [PMID: 34659510 PMCID: PMC8515564 DOI: 10.3892/etm.2021.10798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Chemoresistance is the primary cause of the poor outcome of glioblastoma multiforme (GBM) therapy. Leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) is involved in the growth and proliferation of several types of cancer, including gastric cancer and ovarian cancer. Therefore, the aim of the present study was to investigate the role of LGR6 in GBM malignancy and chemoresistance. Cell counting kit-8 and Matrigel®-Transwell assays were conducted to assess GBM cell viability and invasion. The effect of LGR6 on cell cycle progression and activation of Akt signaling was analyzed by performing propidium iodide staining and western blotting, respectively. The results demonstrated that LGR6, a microRNA-1236-3p target candidate, promoted GBM cell viability and invasion, and mediated temozolomide sensitivity in SHG-44 and U251 GBM cells. In addition, LGR6 triggered the activation of the Akt signaling pathway during GBM progression. Collectively, the results of the present study suggested that LGR6 promoted GBM malignancy and chemoresistance, at least in part, by activating the Akt signaling pathway. The results may aid with the identification of a novel therapeutic target and strategy for GBM.
Collapse
Affiliation(s)
- Yuan Yuan Cheng
- Department of Oncology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xue Yang
- Department of Oncology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xin Gao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Si Xin Song
- Department of Neurosurgery, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| | - Ming Feng Yang
- Institute of Basic Medicine of Shangdong, First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| | - Fang Min Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| |
Collapse
|
49
|
Yue Z, Niu X, Yuan Z, Qin Q, Jiang W, He L, Gao J, Ding Y, Liu Y, Xu Z, Li Z, Yang Z, Li R, Xue X, Gao Y, Yue F, Zhang XHF, Hu G, Wang Y, Li Y, Chen G, Siwko S, Gartland A, Wang N, Xiao J, Liu M, Luo J. RSPO2/RANKL-LGR4 signaling regulates osteoclastic pre-metastatic niche formation and bone metastasis. J Clin Invest 2021; 132:144579. [PMID: 34847079 PMCID: PMC8759794 DOI: 10.1172/jci144579] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Therapeutics targeting osteoclasts are commonly used treatments for bone metastasis; however, whether and how osteoclasts regulate pre-metastatic niche and bone tropism is largely unknown. In this study, we report that osteoclast precursors (OPs) can function as a pre-metastatic niche component that facilitates breast cancer (BCa) bone metastasis at early stages. At the molecular level, unbiased GPCR ligand/agonist screening in BCa cells suggested that R-spondin 2 (RSPO2) and RANKL, through interacting with their receptor LGR4, promoted osteoclastic pre-metastatic niche formation and enhanced BCa bone metastasis. This was achieved by RSPO2/RANKL-LGR4 signal modulating WNT inhibitor DKK1 through Gαq and β-catenin signaling. DKK1 directly facilitated OP recruitment through suppressing its receptor low-density lipoprotein-related receptors 5 (LRP5) but not LRP6, upregulating Rnasek expression via inhibiting canonical WNT signaling. In clinical samples, RSPO2, LGR4 and DKK1 expression showed positive correlation with BCa bone metastasis. Furthermore, soluble LGR4 extracellular domain (ECD) protein, acting as a decoy receptor for RSPO2 and RANKL, significantly alleviated bone metastasis and osteolytic lesions in mouse bone metastasis model. These findings provide unique insights into the functional role of OPs as key components of pre-metastatic niche for BCa bone metastasis, indicate RSPO2/RANKL-LGR4 signaling as a promising target for inhibiting BCa bone metastasis.
Collapse
Affiliation(s)
- Zhiying Yue
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Niu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zengjin Yuan
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qin Qin
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenhao Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Jingduo Gao
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Ding
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yanxi Liu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ziwei Xu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhenxi Li
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhengfeng Yang
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Rong Li
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiwen Xue
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yankun Gao
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Fei Yue
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Yi Wang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Geng Chen
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Stefan Siwko
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, United States of America
| | - Alison Gartland
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Jianru Xiao
- Department of Orthopaedic Oncology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Mingyao Liu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jian Luo
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
50
|
Naert T, Çiçek Ö, Ogar P, Bürgi M, Shaidani NI, Kaminski MM, Xu Y, Grand K, Vujanovic M, Prata D, Hildebrandt F, Brox T, Ronneberger O, Voigt FF, Helmchen F, Loffing J, Horb ME, Willsey HR, Lienkamp SS. Deep learning is widely applicable to phenotyping embryonic development and disease. Development 2021; 148:273338. [PMID: 34739029 PMCID: PMC8602947 DOI: 10.1242/dev.199664] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Genome editing simplifies the generation of new animal models for congenital disorders. However, the detailed and unbiased phenotypic assessment of altered embryonic development remains a challenge. Here, we explore how deep learning (U-Net) can automate segmentation tasks in various imaging modalities, and we quantify phenotypes of altered renal, neural and craniofacial development in Xenopus embryos in comparison with normal variability. We demonstrate the utility of this approach in embryos with polycystic kidneys (pkd1 and pkd2) and craniofacial dysmorphia (six1). We highlight how in toto light-sheet microscopy facilitates accurate reconstruction of brain and craniofacial structures within X. tropicalis embryos upon dyrk1a and six1 loss of function or treatment with retinoic acid inhibitors. These tools increase the sensitivity and throughput of evaluating developmental malformations caused by chemical or genetic disruption. Furthermore, we provide a library of pre-trained networks and detailed instructions for applying deep learning to the reader's own datasets. We demonstrate the versatility, precision and scalability of deep neural network phenotyping on embryonic disease models. By combining light-sheet microscopy and deep learning, we provide a framework for higher-throughput characterization of embryonic model organisms. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Thomas Naert
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Özgün Çiçek
- Department of Computer Science, Albert-Ludwigs-University, Freiburg 79100, Germany
| | - Paulina Ogar
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Max Bürgi
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Nikko-Ideen Shaidani
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Michael M Kaminski
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 10115, Germany.,Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Yuxiao Xu
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Kelli Grand
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Marko Vujanovic
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Daniel Prata
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Friedhelm Hildebrandt
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115,USA
| | - Thomas Brox
- Department of Computer Science, Albert-Ludwigs-University, Freiburg 79100, Germany
| | - Olaf Ronneberger
- Department of Computer Science, Albert-Ludwigs-University, Freiburg 79100, Germany.,BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University, Freiburg, Germany.,DeepMind, London WC2H 8AG , UK
| | - Fabian F Voigt
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Zurich 8057, Switzerland; Neuroscience Center Zurich, Zurich 8057, Switzerland
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Zurich 8057, Switzerland; Neuroscience Center Zurich, Zurich 8057, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| | - Marko E Horb
- National Xenopus Resource and Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Soeren S Lienkamp
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland; Swiss National Centre of Competence in Research (NCCR) Kidney Control of Homeostasis (Kidney.CH), Zurich 8057, Switzerland
| |
Collapse
|