1
|
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
|
2
|
Lee SH, Platt S, Lim CH, Ito M, Myung P. The development of hair follicles and nail. Dev Biol 2024; 513:3-11. [PMID: 38759942 DOI: 10.1016/j.ydbio.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
The hair follicle and nail unit develop and regenerate through epithelial-mesenchymal interactions. Here, we review some of the key signals and molecular interactions that regulate mammalian hair follicle and nail formation during embryonic development and how these interactions are reutilized to promote their regeneration during adult homeostasis and in response to skin wounding. Finally, we highlight the role of some of these signals in mediating human hair follicle and nail conditions.
Collapse
Affiliation(s)
- Soung-Hoon Lee
- The Ronald O. Perelman Department of Dermatology and Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Sarah Platt
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology and Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology and Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Peggy Myung
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Li XC, Wang W, Jiang C, Chen YL, Chen JH, Zhang ZW, Luo SJ, Chen RC, Mo PF, Zhong ML, Shi JY, Huang CM, Chen Q, Wu YH. CD206 + M2-like macrophages protect against intervertebral disc degeneration partially by targeting R-spondin-2. Osteoarthritis Cartilage 2024; 32:66-81. [PMID: 37802465 DOI: 10.1016/j.joca.2023.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVE This study aimed to explore the specific function of M2 macrophages in intervertebral disc degeneration (IDD). METHODS Intervertebral disc (IVD) samples from normal (n = 4) and IDD (n = 6) patients were collected, and the expression of M2-polarized macrophage marker, CD206, was investigated using immunohistochemical staining. Nucleus pulposus cells (NPCs) in a TNF-α environment were obtained, and a mouse caudal IVD puncture model was established. Mice with Rheb deletions, specifically in the myeloid lineage, were generated and subjected to surgery-induced IDD. IDD-induced damage and cell apoptosis were measured using histological scoring, X-ray imaging, immunohistochemical staining, and TdT-mediated dUTP nick end labeling (TUNEL) assay. Finally, mice and NPCs were treated with R-spondin-2 (Rspo2) or anti-Rspo2 to investigate the role of Rspo2 in IDD. RESULTS Accumulation of CD206 in human and mouse IDD tissues was detected. Rheb deletion in the myeloid lineage (RheBcKO) increased the number of CD206+ M2-like macrophages (mean difference 18.6% [15.7-21.6%], P < 0.001), decreased cell apoptosis (mean difference -15.6% [-8.9 to 22.2%], P = 0.001) and attenuated the IDD process in the mouse IDD model. NPCs treated with Rspo2 displayed increased extracellular matrix catabolism and apoptosis; co-culture with a conditioned medium derived from RheBcKO mice inhibited these changes. Anti-Rspo2 treatment in the mouse caudal IVD puncture model exerted protective effects against IDD. CONCLUSIONS Promoting CD206+ M2-like macrophages could reduce Rspo2 secretion, thereby alleviating experimental IDD. Rheb deletion may help M2-polarized macrophages accumulate and attenuate experimental IDD partially by inhibiting Rspo2 production. Hence, M2-polarized macrophages and Rspo2 may serve as therapeutic targets for IDD.
Collapse
Affiliation(s)
- Xiao-Chuan Li
- Department of Orthopedic Surgery, Gaozhou People's Hospital, No.89 XiGuan Rd, Gaozhou 525200, Guangdong, China; Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Postdoctoral Innovation Practice Base of Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China
| | - Wei Wang
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang 524023, Guangdong, China
| | - Cheng Jiang
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang 524023, Guangdong, China
| | - Yong-Long Chen
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang 524023, Guangdong, China
| | - Jiong-Hui Chen
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang 524023, Guangdong, China
| | - Zhen-Wu Zhang
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China; Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang 524023, Guangdong, China
| | - Shao-Jian Luo
- Department of Orthopedic Surgery, Gaozhou People's Hospital, No.89 XiGuan Rd, Gaozhou 525200, Guangdong, China
| | - Rong-Chun Chen
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China
| | - Ping-Fan Mo
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China
| | - Ming-Liang Zhong
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China
| | - Jiang-You Shi
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China
| | - Chun-Ming Huang
- Department of Orthopedic Surgery, Gaozhou People's Hospital, No.89 XiGuan Rd, Gaozhou 525200, Guangdong, China; Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou 525200, China.
| | - Qin Chen
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China.
| | - Yao-Hong Wu
- Department of Spinal Surgery, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou 341000, Jiangxi, China.
| |
Collapse
|
4
|
Wu G, Wang D, Xiong F, Liu W, Wang Q, Chen J, Wang B, Chen Y. Upregulation of RSPO3 via targeted promoter DNA demethylation inhibits the progression of cholangiocarcinoma. Clin Epigenetics 2023; 15:177. [PMID: 37932819 PMCID: PMC10629118 DOI: 10.1186/s13148-023-01592-9] [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: 08/07/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) refers to a collection of malignant tumors that develop from the biliary epithelium. Extensive clinical evidence and epidemiological observations indicate a concerning increase in both the incidence and mortality rates of CCA. Surgical resection is currently the sole available cure for CCA. However, it is unfortunate that only a fraction of patients has access to surgery at the time of diagnosis. Moreover, there is a high incidence of cancer recurrence after resection, and systemic treatments have limited efficacy. Therefore, the identification of novel biomarkers for CCA-targeted molecular therapy remains a crucial task in oncology research. RESULTS Our study demonstrated that low expression of RSPO3 was associated with poorer survival rates in patients with CCA. We found that the RSPO3 promoter DNA was hypermethylated in CCA, which was correlated with the low expression of RSPO3. The expression of RSPO3 was influenced by the balance between the DNA methyltransferase DNMT3a and the DNA demethylase TET1 in CCA. In vitro and in vivo experiments showed that targeting RSPO3 promoter DNA methylation using dCas9DNMT3a promoted tumorigenicity of CCA, while targeted RSPO3 promoter DNA demethylation using dCas9TET1CD inhibited CCA tumorigenicity. Additionally, in our primary CCA model, knockdown of Rspo3 promoted CCA progression, whereas overexpression of Rspo3 inhibited CCA progression. CONCLUSIONS Our findings suggest that increased methylation and decreased expression of RSPO3 may indicate a poor prognosis in CCA. Restoring RSPO3 expression by targeting promoter DNA demethylation could offer insights for precise treatment of CCA.
Collapse
Affiliation(s)
- Guanhua Wu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Da Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Fei Xiong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Wenzheng Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Qi Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Junsheng Chen
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Bing Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China.
| | - Yongjun Chen
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China.
| |
Collapse
|
5
|
Doherty L, Wan M, Peterson A, Youngstrom DW, King JS, Kalajzic I, Hankenson KD, Sanjay A. Wnt-associated adult stem cell marker Lgr6 is required for osteogenesis and fracture healing. Bone 2023; 169:116681. [PMID: 36708855 PMCID: PMC10015414 DOI: 10.1016/j.bone.2023.116681] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Despite the remarkable regenerative capacity of skeletal tissues, nonunion of bone and failure of fractures to heal properly presents a significant clinical concern. Stem and progenitor cells are present in bone and become activated following injury; thus, elucidating mechanisms that promote adult stem cell-mediated healing is important. Wnt-associated adult stem marker Lgr6 is implicated in the regeneration of tissues with well-defined stem cell niches in stem cell-reliant organs. Here, we demonstrate that Lgr6 is dynamically expressed in osteoprogenitors in response to fracture injury. We used an Lgr6-null mouse model and found that Lgr6 expression is necessary for maintaining bone volume and efficient postnatal bone regeneration in adult mice. Skeletal progenitors isolated from Lgr6-null mice have reduced colony-forming potential and reduced osteogenic differentiation capacity due to attenuated cWnt signaling. Lgr6-null mice consist of a lower proportion of self-renewing stem cells. In response to fracture injury, Lgr6-null mice have a deficiency in the proliferation of periosteal progenitors and reduced ALP activity. Further, analysis of the bone regeneration phase and remodeling phase of fracture healing in Lgr6-null mice showed impaired endochondral ossification and decreased mineralization. We propose that in contrast to not being required for successful skeletal development, Lgr6-positive cells have a direct role in endochondral bone repair.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA; School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Anna Peterson
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Justin S King
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Ivo Kalajzic
- School of Dental Medicine, UConn Health, Farmington, CT 06030, USA; Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA.
| |
Collapse
|
6
|
Gill AK, McCormick PJ, Sochart D, Nalesso G. Wnt signalling in the articular cartilage: A matter of balance. Int J Exp Pathol 2023; 104:56-63. [PMID: 36843204 PMCID: PMC10009303 DOI: 10.1111/iep.12472] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/28/2023] Open
Abstract
Degradation of the articular cartilage is a hallmark of osteoarthritis, a progressive and chronic musculoskeletal condition, affecting millions of people worldwide. The activation of several signalling cascades is altered during disease development: among them, the Wnt signalling plays a pivotal role in the maintenance of tissue homeostasis. Increasing evidence is showing that its activation needs to be maintained within a certain range to avoid the triggering of degenerative mechanisms. In this review, we summarise our current knowledge about how a balanced activation of the Wnt signalling is maintained in the articular cartilage, with a particular focus on receptor-mediated mechanisms.
Collapse
Affiliation(s)
- Amandeep Kaur Gill
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary, University of London, London, UK
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary, University of London, London, UK
| | - David Sochart
- South West London Elective Orthopaedic Centre, Epsom, UK
| | - Giovanna Nalesso
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, UK
| |
Collapse
|
7
|
Xu S, Yang X, Chen Q, Liu Z, Chen Y, Yao X, Xiao A, Tian J, Xie L, Zhou M, Hu Z, Zhu F, Xu X, Hou F, Nie J. Leukemia inhibitory factor is a therapeutic target for renal interstitial fibrosis. EBioMedicine 2022; 86:104312. [PMID: 36335669 PMCID: PMC9646860 DOI: 10.1016/j.ebiom.2022.104312] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The role of the IL6 family members in organ fibrosis, including renal interstitial fibrosis (TIF), has been widely explored. However, few studies have ever simultaneously examined them in the same cohort of patients. Besides, the role of leukemia inhibitory factor (LIF) in TIF remains unclear. METHODS RNA-seq data of kidney biopsies from chronic kidney disease (CKD) patients, in both public databases and our assays, were used to analyze transcript levels of IL6 family members. Two TIF mouse models, the unilateral ureteral obstruction (UUO) and the ischemia reperfusion injury (IRI), were employed to validate the finding. To assess the role of LIF in vivo, short hairpin RNA, lenti-GFP-LIF was used to knockdown LIF receptor (LIFR), overexpress LIF, respectively. LIF-neutralizing antibody was used in therapeutic studies. Whether urinary LIF could be used as a promising predictor for CKD progression was investigated in a prospective observation patient cohort. FINDINGS Among IL6 family members, LIF is the most upregulated one in both human and mouse renal fibrotic lesions. The mRNA level of LIF negatively correlated with eGFR with the strongest correlation and the smallest P value. Baseline urinary concentrations of LIF in CKD patients predict the risk of CKD progression to end-stage kidney disease by Kaplan-Meier analysis. In mouse TIF models, knockdown of LIFR alleviated TIF; conversely, overexpressing LIF exacerbated TIF. Most encouragingly, visible efficacy against TIF was observed by administering LIF-neutralizing antibodies to mice. Mechanistically, LIF-LIFR-EGR1 axis and Sonic Hedgehog signaling formed a vicious cycle between fibroblasts and proximal tubular cells to augment LIF expression and promote the pro-fibrotic response via ERK and STAT3 activation. INTERPRETATION This study discovered that LIF is a noninvasive biomarker for the progression of CKD and a potential therapeutic target of TIF. FUNDINGS Stated in the Acknowledgements section of the manuscript.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Fanfan Hou
- Corresponding author. Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jing Nie
- Corresponding author. Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Nelson AL, Fontana G, Miclau E, Rongstad M, Murphy W, Huard J, Ehrhart N, Bahney C. Therapeutic approaches to activate the canonical Wnt pathway for bone regeneration. J Tissue Eng Regen Med 2022; 16:961-976. [PMID: 36112528 PMCID: PMC9826348 DOI: 10.1002/term.3349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/05/2022] [Accepted: 09/01/2022] [Indexed: 01/12/2023]
Abstract
Activation of the canonical Wingless-related integration site (Wnt) pathway has been shown to increase bone formation and therefore has therapeutic potential for use in orthopedic conditions. However, attempts at developing an effective strategy to achieve Wnt activation has been met with several challenges. The inherent hydrophobicity of Wnt ligands makes isolating and purifying the protein difficult. To circumvent these challenges, many have sought to target extracellular inhibitors of the Wnt pathway, such as Wnt signaling pathway inhibitors Sclerostin and Dickkopf-1, or to use small molecules, ions and proteins to increase target Wnt genes. Here, we review systemic and localized bioactive approaches to enhance bone formation or improve bone repair through antibody-based therapeutics, synthetic Wnt surrogates and scaffold doping to target canonical Wnt. We conclude with a brief review of emerging technologies, such as mRNA therapy and Clustered Regularly Interspaced Short Palindromic Repeats technology, which serve as promising approaches for future clinical translation.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA
| | - GianLuca Fontana
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Elizabeth Miclau
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA
| | - Mallory Rongstad
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - William Murphy
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA,Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Johnny Huard
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Nicole Ehrhart
- School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Chelsea Bahney
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA,Orthopaedic Trauma InstituteUniversity of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
| |
Collapse
|
9
|
Lu T, Forgetta V, Greenwood CMT, Richards JB. Identifying Causes of Fracture Beyond Bone Mineral Density: Evidence From Human Genetics. J Bone Miner Res 2022; 37:1592-1602. [PMID: 35689460 DOI: 10.1002/jbmr.4632] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 05/28/2022] [Accepted: 06/04/2022] [Indexed: 11/10/2022]
Abstract
New therapies may help to prevent osteoporotic fractures other than through increasing bone mineral density (BMD). Because fracture risk has an important genetic component, we aim to identify loci increasing fracture risk that do not decrease BMD, using a recently-proposed structural equation model adapted to remove genetic influences of BMD on fracture risk. We used summary statistics of the largest genome-wide association studies (GWASs) for BMD and for fracture in these analyses. We next estimated the genetic correlation between the non-BMD or BMD-related genetic effects and other clinical risk factors for fracture. Last, based on white British participants in the UK Biobank, we conducted genetic risk score analyses to assess whether the aggregated genetic effects conferred increased major osteoporotic fracture risk. We found that only three loci affecting fracture risk exhibited genetic effects not mediated by BMD: SOST, CPED1-WNT16, and RSPO3, while these three loci simultaneously conferred BMD-related effects. No strong genetic associations between non-BMD or BMD-related effects and 16 clinical risk factors were observed. However, non-BMD effects might be genetic correlated with hip bone size. In the UK Biobank, a 1 standard deviation (1-SD) increase in the non-BMD genetic risk score conferred an odds ratio of 1.17 for incident major osteoporotic fracture, compared to 1.29 by a BMD-related genetic risk score. Our study suggests that the majority of common genetic predisposition toward fracture risk acts upon BMD. Although non-BMD genetic effects may exist, they are not strongly correlated with most traditional clinical risk factors. Risk loci harboring non-BMD genetic effects may influence other perspectives of bone quality, or confer effects that existing GWASs fail to capture, but they demonstrate weaker impact on fracture risk than BMD-related genetic effects. These findings suggest that most successful drug development programs for osteoporosis should focus on pathways identified through BMD-associated loci. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada.,Quantitative Life Sciences Program, McGill University, Montreal, Canada
| | - Vincenzo Forgetta
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada
| | - J Brent Richards
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada.,Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 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
|
11
|
Hao1 Is Not a Pathogenic Factor for Ectopic Ossifications but Functions to Regulate the TCA Cycle In Vivo. Metabolites 2022; 12:metabo12010082. [PMID: 35050204 PMCID: PMC8780519 DOI: 10.3390/metabo12010082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/01/2023] Open
Abstract
Ossification of the posterior longitudinal ligament (OPLL), a disease characterized by the ectopic ossification of a spinal ligament, promotes neurological disorders associated with spinal canal stenosis. While blocking ectopic ossification is mandatory to prevent OPLL development and progression, the mechanisms underlying the condition remain unknown. Here we show that expression of hydroxyacid oxidase 1 (Hao1), a gene identified in a previous genome-wide association study (GWAS) as an OPLL-associated candidate gene, specifically and significantly decreased in fibroblasts during osteoblast differentiation. We then newly established Hao1-deficient mice by generating Hao1-flox mice and crossing them with CAG-Cre mice to yield global Hao1-knockout (CAG-Cre/Hao1flox/flox; Hao1 KO) animals. Hao1 KO mice were born normally and exhibited no obvious phenotypes, including growth retardation. Moreover, Hao1 KO mice did not exhibit ectopic ossification or calcification. However, urinary levels of some metabolites of the tricarboxylic acid (TCA) cycle were significantly lower in Hao1 KO compared to control mice based on comprehensive metabolomic analysis. Our data indicate that Hao1 loss does not promote ectopic ossification, but rather that Hao1 functions to regulate the TCA cycle in vivo.
Collapse
|
12
|
Kang M, Ahn B, Youk S, Lee YM, Kim JJ, Ha JH, Park C. Tracing the Origin of the RSPO2 Long-Hair Allele and Epistatic Interaction between FGF5 and RSPO2 in Sapsaree Dog. Genes (Basel) 2022; 13:genes13010102. [PMID: 35052442 PMCID: PMC8775186 DOI: 10.3390/genes13010102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Genetic analysis of the hair-length of Sapsaree dogs, a Korean native dog breed, showed a dominant mode of inheritance for long hair. Genome-Wide Association Study (GWAS) analysis and subsequent Mendelian segregation analysis revealed an association between OXR1, RSPO2, and PKHD1L1 on chromosome 13 (CFA13). We identified the previously reported 167 bp insertion in RSPO2 3’ untranslated region as a causative mutation for hair length variations. The analysis of 118 dog breeds and wolves revealed the selection signature on CFA13 in long-haired breeds. Haplotype analysis showed the association of only a few specific haplotypes to the breeds carrying the 167 bp insertion. The genetic diversity in the neighboring region linked to the insertion was higher in Sapsarees than in other Asian and European dog breeds carrying the same variation, suggesting an older history of its insertion in the Sapsaree genome than in that of the other breeds analyzed in this study. Our results show that the RSPO2 3’ UTR insertion is responsible for not only the furnishing phenotype but also determining the hair length of the entire body depending on the genetic background, suggesting an epistatic interaction between FGF5 and RSPO2 influencing the hair-length phenotype in dogs.
Collapse
Affiliation(s)
- Mingue Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (M.K.); (B.A.); (S.Y.)
| | - Byeongyong Ahn
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (M.K.); (B.A.); (S.Y.)
| | - Seungyeon Youk
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (M.K.); (B.A.); (S.Y.)
| | - Yun-Mi Lee
- Department of Biotechnology, Yeungnam University, Gyeongsan 36461, Korea; (Y.-M.L.); (J.-J.K.)
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 36461, Korea; (Y.-M.L.); (J.-J.K.)
| | - Ji-Hong Ha
- Korean Sapsaree Foundation, Gyeongsan 38412, Korea;
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (M.K.); (B.A.); (S.Y.)
- Correspondence: ; Tel.: +82-10-8826-1363
| |
Collapse
|
13
|
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: 2.0] [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
|
14
|
Garcia J, Smith SS, Karki S, Drissi H, Hrdlicka HH, Youngstrom DW, Delany AM. miR-433-3p suppresses bone formation and mRNAs critical for osteoblast function in mice. J Bone Miner Res 2021; 36:1808-1822. [PMID: 34004029 DOI: 10.1002/jbmr.4339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) are key posttranscriptional regulators of osteoblastic commitment and differentiation. miR-433-3p was previously shown to target Runt-related transcription factor 2 (Runx2) and to be repressed by bone morphogenetic protein (BMP) signaling. Here, we show that miR-433-3p is progressively decreased during osteoblastic differentiation of primary mouse bone marrow stromal cells in vitro, and we confirm its negative regulation of this process. Although repressors of osteoblastic differentiation often promote adipogenesis, inhibition of miR-433-3p did not affect adipocyte differentiation in vitro. Multiple pathways regulate osteogenesis. Using luciferase-3' untranslated region (UTR) reporter assays, five novel miR-433-3p targets involved in parathyroid hormone (PTH), mitogen-activated protein kinase (MAPK), Wnt, and glucocorticoid signaling pathways were validated. We show that Creb1 is a miR-433-3p target, and this transcription factor mediates key signaling downstream of PTH receptor activation. We also show that miR-433-3p targets hydroxysteroid 11-β dehydrogenase 1 (Hsd11b1), the enzyme that locally converts inactive glucocorticoids to their active form. miR-433-3p dampens glucocorticoid signaling, and targeting of Hsd11b1 could contribute to this phenomenon. Moreover, miR-433-3p targets R-spondin 3 (Rspo3), a leucine-rich repeat-containing G-protein coupled receptor (LGR) ligand that enhances Wnt signaling. Notably, Wnt canonical signaling is also blunted by miR-433-3p activity. In vivo, expression of a miR-433-3p inhibitor or tough decoy in the osteoblastic lineage increased trabecular bone volume. Mice expressing the miR-433-3p tough decoy displayed increased bone formation without alterations in osteoblast or osteoclast numbers or surface, indicating that miR-433-3p decreases osteoblast activity. Overall, we showed that miR-433-3p is a negative regulator of bone formation in vivo, targeting key bone-anabolic pathways including those involved in PTH signaling, Wnt, and endogenous glucocorticoids. Local delivery of miR-433-3p inhibitor could present a strategy for the management of bone loss disorders and bone defect repair. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- John Garcia
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Spenser S Smith
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University and Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Henry H Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Daniel W Youngstrom
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
15
|
Wang Z, Wang Y, Ma X, Dang C. RSPO2 silence inhibits tumorigenesis of nasopharyngeal carcinoma by ZNRF3/Hedgehog-Gli1 signal pathway. Life Sci 2021; 282:119817. [PMID: 34273374 DOI: 10.1016/j.lfs.2021.119817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/27/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
R-spondins 2 (RSPO2) protein is a member of RSPO family which plays an essential role in stem cell survival, development and tumorigenicity. There has several evidence suggested that RSPO2 involved in breast, gastric, liver and colorectal cancer. However, the specific function and mechanism of RSPO2 in nasopharyngeal carcinoma (NPC) remain unknown. In the present study, we first observed that RSPO2 expression was elevated in NPC cell lines SUNE-6-10B, SUNE-5-8F, and CNE-1 compared with the normal laryngeal epithelia cell line NP69. Knockdown of RSPO2 significantly inhibits SUNE-6-10B and CNE-1 cell survival and proliferation by using CCK-8 assay and Edu assay, respectively. Further studies verified that RSPO2 silence suppressed migration and invasion of SUNE-6-10B and CNE-1 cells. Further studies suggested that RSPO2 silence suppressed epithelial-to-mesenchymal transition (EMT) related protein E-cadherin expression and promoted Vimentin and N-cadherin expression both in SUNE-6-10B and CNE-1 cells. Molecular mechanism explorations showed that RSPO2 deletion increased ZNRF3 expression and inhibited Gli1 expression. Additionally, knockdown ZNRF3 expression or overexpression Gli1 both reversed the effects of RSPO2 silence on NPC growth and metastasis. Finally, RSPO2 depletion was impaired NPC tumor growth in vivo animal experiments. In conclusion, the present study confirmed that RSPO2 silence inhibits the tumorigenesis of NPC via ZNRF3/Hedgehog-Gli1 signal pathway.
Collapse
Affiliation(s)
- ZhongWei Wang
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - YaLi Wang
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - XiuLong Ma
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - ChengXue Dang
- Tumor Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
16
|
Chen X, Chen L, Tan J, Zhang L, Xia J, Cheng B, Zhang W. Rspo1-LGR4 axis in BMSCs protects bone against radiation-induced injury through the mTOR-dependent autophagy pathway. J Cell Physiol 2021; 236:4273-4289. [PMID: 33452710 DOI: 10.1002/jcp.30051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/12/2023]
Abstract
While mesenchymal stem cells (MSCs) have been widely used to repair radiation-induced bone damage, the molecular mechanism underlying the effects of MSCs in the maintenance of bone homeostasis under radiation stress remains largely unknown. In this study, the role and mechanisms of R-spondin 1 (Rspo1)-leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis on the initiation of self-defense of bone mesenchymal stem cells (BMSCs) and maintenance of bone homeostasis under radiation stress were investigated. Interestingly, radiation increased levels of Rspo1 and LGR4 in BMSCs. siRNA knockdown of Rspo1 or LGR4 aggravated radiation-induced impairment of self-renewal ability and osteogenic differentiation potential of BMSCs. However, exogenous Rspo1 significantly attenuated radiation-induced depletion of BMSCs, and promoted the lineage shift towards osteoblasts. This alteration was associated with the reversal of mammalian target of rapamycin (mTOR) activation and autophagy decrement. Pharmacological and genetic blockade of autophagy attenuated the radio-protective effects of Rspo1, rendering BMSCs more vulnerable to radiation-induced injury. Then bone radiation injury was induced in C57BL6J mice to further determine the radio-protective effects of Rspo1. In mice, administration of Rspo1 recombinant protein alleviated radiation-induced bone loss. Our results uncover that Rspo1-LGR4-mTOR-autophagy axis are key mechanisms by which BMSCs initiate self-defense against radiation and maintain bone homeostasis. Targeting Rspo1-LGR4 may provide a novel strategy for the intervention of radiation-induced bone damage.
Collapse
Affiliation(s)
- Xiaodan Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lingling Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jiali Tan
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liping Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weizhen Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, China
| |
Collapse
|
17
|
Alhazmi N, Carroll SH, Kawasaki K, Woronowicz KC, Hallett SA, Macias Trevino C, Li EB, Baron R, Gori F, Yelick PC, Harris MP, Liao EC. Synergistic roles of Wnt modulators R-spondin2 and R-spondin3 in craniofacial morphogenesis and dental development. Sci Rep 2021; 11:5871. [PMID: 33712657 PMCID: PMC7954795 DOI: 10.1038/s41598-021-85415-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 02/26/2021] [Indexed: 12/01/2022] Open
Abstract
Wnt signaling plays a critical role in craniofacial patterning, as well as tooth and bone development. Rspo2 and Rspo3 are key regulators of Wnt signaling. However, their coordinated function and relative requirement in craniofacial development and odontogensis are poorly understood. We showed that in zebrafish rspo2 and rspo3 are both expressed in osteoprogenitors in the embryonic craniofacial skeleton. This is in contrast to mouse development, where Rspo3 is expressed in osteoprogenitors while Rspo2 expression is not observed. In zebrafish, rspo2 and rspo3 are broadly expressed in the pulp, odontoblasts and epithelial crypts. However, in the developing molars of the mouse, Rspo3 is largely expressed in the dental follicle and alveolar mesenchyme while Rspo2 expression is restricted to the tooth germ. While Rspo3 ablation in the mouse is embryonic lethal, zebrafish rspo3-/- mutants are viable with modest decrease in Meckel's cartilage rostral length. However, compound disruption of rspo3 and rspo2 revealed synergistic roles of these genes in cartilage morphogenesis, fin development, and pharyngeal tooth development. Adult rspo3-/- zebrafish mutants exhibit a dysmorphic cranial skeleton and decreased average tooth number. This study highlights the differential functions of Rspo2 and Rspo3 in dentocranial morphogenesis in zebrafish and in mouse.
Collapse
Affiliation(s)
- Nora Alhazmi
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Shannon H Carroll
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kenta Kawasaki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
| | - Katherine C Woronowicz
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Orthopedics, Boston Children's Hospital, Boston, MA, USA
| | - Shawn A Hallett
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Claudio Macias Trevino
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Edward B Li
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Roland Baron
- Harvard School of Dental Medicine, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Pamela C Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Matthew P Harris
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Orthopedics, Boston Children's Hospital, Boston, MA, USA
| | - Eric C Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Shriners Hospital for Children, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Kaur S, Roberts DD. Differential intolerance to loss of function and missense mutations in genes that encode human matricellular proteins. J Cell Commun Signal 2021; 15:93-105. [PMID: 33415696 PMCID: PMC7904989 DOI: 10.1007/s12079-020-00598-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Targeted gene disruption in mice has provided valuable insights into the functions of matricellular proteins. Apart from missense and loss of function mutations that have been associated with inherited diseases, however, their functions in humans remain unclear. The availability of deep exome sequencing data from over 140,000 individuals in the Genome Aggregation Database provided an opportunity to examine intolerance to loss of function and missense mutations in human matricellular genes. The probability of loss-of-function intolerance (pLI) differed widely within members of the thrombospondin, CYR61/CTGF/NOV (CCN), tenascin, small integrin-binding ligand N-linked glycoproteins (SIBLING), and secreted protein, acidic and rich in cysteine (SPARC) gene families. Notably, pLI values in humans had limited correlation with viability of the corresponding homozygous null mice. Among the thrombospondins, only THBS1 was highly loss-intolerant (pLI = 1). In contrast, Thbs1 is not essential for viability in mice. Several known thrombospondin-1 receptors were similarly loss-intolerant, although thrombospondin-1 is not the exclusive ligand for some of these receptors. The frequencies of missense mutations in THBS1 and the gene encoding its signaling receptor CD47 indicated conservation of some residues implicated in specific receptor binding. Deficits in missense mutations were also observed for other thrombospondin genes and for SPARC, SPOCK1, SPOCK2, TNR, and DSPP. The intolerance of THBS1 to loss of function in humans and elevated pLI values for THBS2, SPARC, SPOCK1, TNR, and CCN1 support important functions for these matricellular protein genes in humans, some of which may relate to functions in reproduction or responding to environmental stresses.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Building 10 Room 2S235, 10 Center Drive MSC1500, Bethesda, MD, 20892-1500, USA.
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Building 10 Room 2S235, 10 Center Drive MSC1500, Bethesda, MD, 20892-1500, USA.
| |
Collapse
|
19
|
Abstract
R-spondin (RSPO) proteins amplify Wnt signaling and stimulate regeneration in a variety of tissues. To repair tissue in a tissue-specific manner, tissue-targeted RSPO mimetic molecules are desired. Here, we mutated RSPO (RSPO2 F105R/F109A) to eliminate LGR binding while preserving ZNRF3/RNF43 binding and targeted the mutated RSPO to a liver specific receptor, ASGR1. The resulting bi-specific molecule (αASGR1-RSPO2-RA) enhanced Wnt signaling effectively in vitro, and its activity was limited to ASGR1 expressing cells. Systemic administration of αASGR1-RSPO2-RA in mice specifically upregulated Wnt target genes and stimulated cell proliferation in liver but not intestine (which is more responsive to non-targeted RSPO2) in healthy mice, and improved liver function in diseased mice. These results not only suggest that a tissue-specific RSPO mimetic protein can stimulate regeneration in a cell-specific manner, but also provide a blueprint of how a tissue-specific molecule might be constructed for applications in a broader context.
Collapse
|
20
|
Doherty L, Sanjay A. LGRs in Skeletal Tissues: An Emerging Role for Wnt-Associated Adult Stem Cell Markers in Bone. JBMR Plus 2020; 4:e10380. [PMID: 32666024 PMCID: PMC7340442 DOI: 10.1002/jbm4.10380] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/18/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeat-containing G protein-coupled receptors (LGRs) are adult stem cell markers that have been described across various stem cell niches, and expression of LGRs and their corresponding ligands (R-spondins) has now been reported in multiple bone-specific cell types. The skeleton harbors elusive somatic stem cell populations that are exceedingly compartment-specific and under tight regulation from various signaling pathways. Skeletal progenitors give rise to multiple tissues during development and during regenerative processes of bone, requiring postnatal endochondral and intramembranous ossification. The relevance of LGRs and the LGR/R-spondin ligand interaction in bone and tooth biology is becoming increasingly appreciated. LGRs may define specific stem cell and progenitor populations and their behavior during both development and regeneration, and their role as Wnt-associated receptors with specific ligands poses these proteins as unique therapeutic targets via potential R-spondin agonism. This review seeks to outline the current literature on LGRs in the context of bone and its associated tissues, and points to key future directions for studying the functional role of LGRs and ligands in skeletal biology. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic SurgeryUConn HealthFarmingtonCTUSA
| | - Archana Sanjay
- Department of Orthopaedic SurgeryUConn HealthFarmingtonCTUSA
| |
Collapse
|
21
|
Bai DP, Chen Y, Hu YQ, He WF, Shi YZ, Fan QM, Luo RT, Li A. Transcriptome analysis of genes related to gonad differentiation and development in Muscovy ducks. BMC Genomics 2020; 21:438. [PMID: 32590948 PMCID: PMC7318502 DOI: 10.1186/s12864-020-06852-z] [Citation(s) in RCA: 12] [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: 12/06/2019] [Accepted: 06/19/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sex-related genes play a crucial role in gonadal differentiation into testes or ovaries. However, the genetic control of gonadal differentiation in Muscovy ducks remains unknown. Therefore, the objective of our study was to screen new candidate genes associated with ovarian and testicular development. RESULTS In this study, 24 males before gonadal differentiation (MB), 24 females before gonadal differentiation (FB), 24 males after gonadal differentiation (MA) and 24 females after gonadal differentiation (FA) were selected from Putian Muscovy ducks, forming 4 groups. RNA-Seq revealed 101.76 Gb of clean reads and 2800 differentially expressed genes (DEGs), including 46 in MB vs FB, 609 in MA vs FA, 1027 in FA vs FB, and 1118 in MA vs MB. A total of 146 signalling pathways were enriched by KEGG analysis, among which 20, 108, 108 and 116 signalling pathways were obtained in MB vs FB, MA vs MB, MA vs FA and FA vs FB, respectively. In further GO and KEGG analyses, a total of 21 candidate genes related to gonad differentiation and development in Muscovy ducks were screened. Among these, 9 genes were involved in the differentiation and development of the testes, and 12 genes were involved in the differentiation and development of the ovaries. In addition, RNA-Seq data revealed 2744 novel genes. CONCLUSIONS RNA-Seq data revealed 21 genes related to gonadal differentiation and development in Muscovy ducks. We further identified 12 genes, namely, WNT5B, HTRA3, RSPO3, BMP3, HNRNPK, NIPBL, CREB3L4, DKK3, UBE2R2, UBPL3KCMF1, ANXA2, and OSR1, involved in the differentiation and development of ovaries. Moreover, 9 genes, namely, TTN, ATP5A1, DMRT1, DMRT3, AMH, MAP3K1, PIK3R1, AGT and ADAMTSL1, were related to the differentiation and development of testes. Moreover, after gonadal differentiation, DMRT3, AMH, PIK3R1, ADAMTSL1, AGT and TTN were specifically highly expressed in males. WNT5B, ANXA2 and OSR1 were specifically highly expressed in females. These results provide valuable information for studies on the sex control of Muscovy ducks and reveal novel candidate genes for the differentiation and development of testes and ovaries.
Collapse
Affiliation(s)
- Ding-Ping Bai
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yue Chen
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yu-Qiong Hu
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Wen-Feng He
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yu-Zhu Shi
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Qin-Ming Fan
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Ru-Tang Luo
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Ang Li
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| |
Collapse
|
22
|
Zhou YM, Yang YY, Jing YX, Yuan TJ, Sun LH, Tao B, Liu JM, Zhao HY. BMP9 Reduces Bone Loss in Ovariectomized Mice by Dual Regulation of Bone Remodeling. J Bone Miner Res 2020; 35:978-993. [PMID: 31914211 DOI: 10.1002/jbmr.3957] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/22/2019] [Accepted: 12/28/2019] [Indexed: 12/18/2022]
Abstract
Bone remodeling is dynamic and is tightly regulated through bone resorption dominated by osteoclasts and bone formation dominated by osteoblasts. Imbalances in this process can cause various pathological conditions, such as osteoporosis. Bone morphogenetic protein 9 (BMP9), a biomolecule produced and secreted by the liver, has many pharmacological effects, including anti-liver fibrosis, antitumor, anti-heart failure, and antidiabetic activities. However, the effects of BMP9 on the regulation of osteoblast and osteoclast functions and the underlying molecular mechanism(s) have not yet been investigated. In this study, BMP9 increased the expression of osteoblastogenic gene markers, such as ALP, Cola1, OCN, RUNX2, and OSX, and ALP activity in MC3T3-E1 cells by upregulating LGR6 and activating the Wnt/β-catenin pathway. BMP9 also suppressed receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation of bone marrow macrophages (BMMs) by inhibiting the Akt-NF-κB-NFATc1 pathway. More importantly, in an ovariectomy (OVX) mouse model, BMP9 attenuated bone loss and improved bone biomechanical properties in vivo by increasing bone-forming activity and suppressing bone resorption activity. Accordingly, our current work highlights the dual regulatory effects that BMP9 exerts on bone remodeling by promoting bone anabolic activity and inhibiting osteoclast differentiation in OVX mice. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yan-Man Zhou
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Yu-Ying Yang
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Yi-Xuan Jing
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Tian-Jiao Yuan
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Li-Hao Sun
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Bei Tao
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Hong-Yan Zhao
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
23
|
Munguía-Reyes A, Balderas-Martínez YI, Becerril C, Checa M, Ramírez R, Ortiz B, Meléndez-Zajgla J, Pardo A, Selman M. R-Spondin-2 Is Upregulated in Idiopathic Pulmonary Fibrosis and Affects Fibroblast Behavior. Am J Respir Cell Mol Biol 2019; 59:65-76. [PMID: 29345973 DOI: 10.1165/rcmb.2017-0115oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the expansion of the myofibroblast population, excessive extracellular matrix accumulation, and destruction of the lung parenchyma. The R-spondin family (RSPO) comprises a group of proteins essential for development. Among them, RSPO2 is expressed primarily in the lungs, and its mutations cause severe defects in the respiratory tract. Interestingly, RSPO2 participates in the canonical Wingless/int1 pathway, a critical route in the pathogenesis of IPF. Thus, the aim of this study was to examine the expression and putative role of RSPO2 in this disease. We found that RSPO2 and its receptor leucine-rich G protein-coupled receptor 6 were upregulated in IPF lungs, where they localized primarily in fibroblasts and epithelial cells. Stimulation of IPF and normal lung fibroblasts with recombinant human RSPO2 resulted in the deregulation of numerous genes, although the transcriptional response was essentially distinct. In IPF fibroblasts, RSPO2 stimulation induced the up- or downregulation of several genes involved in the Wingless/int1 pathway (mainly from noncanonical signaling). In both normal and IPF fibroblasts, RSPO2 modifies the expression of genes implicated in several pathways, including the cell cycle and apoptosis. In accordance with gene expression, the stimulation of normal and IPF fibroblasts with RSPO2 significantly reduced cell proliferation and induced cell death. RSPO2 also inhibited collagen production and increased the expression of matrix metalloproteinase 1. Silencing RSPO2 with shRNA induced the opposite effects. Our findings demonstrate, for the first time to our knowledge, that RSPO2 is upregulated in IPF, where it appears to have an antifibrotic role.
Collapse
Affiliation(s)
- Adrián Munguía-Reyes
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Yalbi I Balderas-Martínez
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico.,2 Cátedra Consejo Nacional de Ciencia y Tecnología (CONACyT)-INER, Mexico City, Mexico
| | - Carina Becerril
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Marco Checa
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Remedios Ramírez
- 3 Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
| | - Blanca Ortiz
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | | | - Annie Pardo
- 3 Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
| | - Moisés Selman
- 1 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| |
Collapse
|
24
|
Liu S, U KP, Zhang J, Tsang LL, Huang J, Tu SP, Jiang X. R-spodin2 enhances canonical Wnt signaling to maintain the stemness of glioblastoma cells. Cancer Cell Int 2018; 18:156. [PMID: 30337838 PMCID: PMC6180579 DOI: 10.1186/s12935-018-0655-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/05/2018] [Indexed: 01/30/2023] Open
Abstract
Background As newly identified Wnt enhancer, R-spondin gene family members have been linked to various cancers; however, their role in isocitrate dehydrogenase-wildtype subtype of human glioblastoma (GBM) cells remains unknown. Methods Human U87 and U251 cell lines were used to perform the experiments. GBM stem-like cells were enriched in stem cell growth media and induced to differentiate using retinoid acid or growth factor deprivation. Wnthigh and Wntlow subpopulations were isolated and evaluated by MTS, sphere formation, transwell migration and xenograft formation assays. Results R-spondin 2 but not R-spondin 3 potentiates Wnt/β-catenin signaling in GBM cell lines. While R-spondin 2 does not affect cell growth, it induces the expression of pluripotent stem cell markers in combination with Wnt3A. GBM stem-like cells are endowed with intrinsic high activity of β-catenin signaling, which can be further intensified by R-spondin 2. In addition, R-spondin2 promotes stem cell self-renewal and suppresses retinoid acid- or growth factor deprivation-induced differentiation, indicating R-spondin 2 maintains stem cell traits in GBM. On the other hand, we identify subpopulations of GBM cells that show distinctive responsiveness to Wnt/β-catenin signaling. Interestingly, Wnthigh and Wntlow cells display distinctive biologic properties. Moreover, Wnthigh cell-inoculated xenografts exhibit enhanced tumorigenicity and increased expression levels of R-spondin 2 compared to Wntlow cell-inoculated xenografts. Conclusion Our study reveals a novel regulatory mechanisms underlying the over-activation of β-catenin-mediated signaling in the pathogenesis of GBM. Electronic supplementary material The online version of this article (10.1186/s12935-018-0655-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Si Liu
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China
| | - Kin Pong U
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China.,2School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Jieting Zhang
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China.,2School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Lai Ling Tsang
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China.,2School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Jiawei Huang
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China
| | - Shui Ping Tu
- 3Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaohua Jiang
- 1Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR People's Republic of China.,2School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| |
Collapse
|
25
|
Arima M, Hasegawa D, Yoshida S, Mitarai H, Tomokiyo A, Hamano S, Sugii H, Wada N, Maeda H. R-spondin 2 promotes osteoblastic differentiation of immature human periodontal ligament cells through the Wnt/β-catenin signaling pathway. J Periodontal Res 2018; 54:143-153. [PMID: 30284717 DOI: 10.1111/jre.12611] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/14/2018] [Accepted: 08/26/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE In this study, we measured the expression of R-spondin 2 (RSPO2) in periodontal ligament (PDL) tissue and cells. Further, we examined the effects of RSPO2 on osteoblastic differentiation of immature human PDL cells (HPDLCs). BACKGROUND R-spondin (RSPO) family proteins are secreted glycoproteins that play important roles in embryonic development and tissue homeostasis through activation of the Wnt/β-catenin signaling pathway. RSPO2, a member of the RSPO family, has been reported to enhance osteogenesis in mice. However, little is known regarding the roles of RSPO2 in PDL tissues. METHODS Expression of RSPO2 in rat PDL tissue and primary HPDLCs was examined by immunohistochemical and immunofluorescence staining, as well as by semiquantitative RT-PCR. The effects of stretch loading on the expression of RSPO2 and Dickkopf-related protein 1 (DKK1) were assessed by quantitative RT-PCR. Expression of receptors for RSPOs, such as Leucine-rich repeat-containing G-protein-coupled receptors (LGRs) 4, 5, and 6 in immature human PDL cells (cell line 2-14, or 2-14 cells), was investigated by semiquantitative RT-PCR. Mineralized nodule formation in 2-14 cells treated with RSPO2 under osteoblastic inductive condition was examined by Alizarin Red S and von Kossa stainings. Nuclear translocation of β-catenin and expression of active β-catenin in 2-14 cells treated with RSPO2 were assessed by immunofluorescence staining and Western blotting analysis, respectively. In addition, the effect of Dickkopf-related protein 1 (DKK1), an inhibitor of Wnt/β-catenin signaling, was also examined. RESULTS Rat PDL tissue and HPDLCs expressed RSPO2, and HPDLCs also expressed RSPO2, while little was found in 2-14 cells. Expression of RSPO2 as well as DKK1 in HPDLCs was significantly upregulated by exposure to stretch loading. LGR4 was predominantly expressed in 2-14 cells, which expressed low levels of LGR5 and LGR6. RSPO2 enhanced the Alizarin Red S and von Kossa-positive reactions in 2-14 cells. In addition, DKK1 suppressed nuclear translocation of β-catenin, activation of β-catenin, and increases of Alizarin Red S and von Kossa-positive reactions in 2-14 cells, all of which were induced by RSPO2 treatment. CONCLUSION RSPO2, which is expressed in PDL tissue and cells, might play an important role in regulating the osteoblastic differentiation of immature human PDL cells through the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Mai Arima
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Daigaku Hasegawa
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Shinichiro Yoshida
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hiromi Mitarai
- Division of General Dentistry, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Atsushi Tomokiyo
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Sayuri Hamano
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Faculty of Dental Science, OBT Research Center, Kyushu University, Fukuoka, Japan
| | - Hideki Sugii
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- Division of General Dentistry, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Skaria T, Bachli E, Schoedon G. RSPO3 impairs barrier function of human vascular endothelial monolayers and synergizes with pro-inflammatory IL-1. Mol Med 2018; 24:45. [PMID: 30157748 PMCID: PMC6116367 DOI: 10.1186/s10020-018-0048-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Endothelial barrier dysfunction characterized by hyperpermeability of the vascular endothelium is a key factor in the pathogenesis of chronic inflammatory diseases and affects clinical outcomes. In states of chronic inflammation, mediators secreted by activated immune cells or vascular endothelium may affect the barrier function and permeability of the vascular endothelium. The matricellular R-spondin family member RSPO3 is produced by inflammatory-activated human monocytes and vascular endothelial cells, but its effects in the regulation of vascular endothelial barrier function remains elusive. METHODS The present study investigates the effects of RSPO3 on the barrier function of adult human primary macro- and micro- vascular endothelial monolayers. Tight monolayers of primary endothelial cells from human coronary and pulmonary arteries, and cardiac, brain, and dermal microvascular beds were treated with RSPO3 either alone or in combination with pro-inflammatory mediator IL-1β. Endothelial barrier function was assessed non-invasively in real-time using Electric Cell-substrate Impedance Sensing. RESULTS RSPO3 treatment critically affected barrier function by enhancing the permeability of all vascular endothelial monolayers investigated. To confer hyperpermeable phenotype in vascular endothelial monolayers, RSPO3 induced inter-endothelial gap formation by disrupting the β-catenin and VE-cadherin alignment at adherens junctions. RSPO3 synergistically enhanced the barrier impairing properties of the pro-inflammatory mediator IL-1β. CONCLUSION Here, we show that the matricellular protein RSPO3 is a mediator of endothelial hyperpermeability that can act in synergy with the inflammatory mediator IL-1β. This finding stimulates further studies to delineate the endothelial barrier impairing properties of RSPO3 and its synergistic interaction with IL-1β in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Tom Skaria
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Esther Bachli
- Department of Medicine, Uster Hospital, Brunnenstrasse 42, CH-8610, Uster, Switzerland
| | - Gabriele Schoedon
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland.
| |
Collapse
|
27
|
Knight MN, Karuppaiah K, Lowe M, Mohanty S, Zondervan RL, Bell S, Ahn J, Hankenson KD. R-spondin-2 is a Wnt agonist that regulates osteoblast activity and bone mass. Bone Res 2018; 6:24. [PMID: 30131881 PMCID: PMC6089978 DOI: 10.1038/s41413-018-0026-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 03/28/2018] [Accepted: 06/26/2018] [Indexed: 12/16/2022] Open
Abstract
The R-spondin family of proteins are Wnt agonists, and the complete embryonic disruption of Rspo2 results in skeletal developmental defects that recapitulate the phenotype observed with Lrp5/6 deficiency. Previous work has shown that R-spondin-2 (Rspo2, RSPO2) is both highly expressed in Wnt-stimulated pre-osteoblasts and its overexpression induces osteoblast differentiation in the same cells, supporting its putative role as a positive autocrine regulator of osteoblastogenesis. However, the role of Rspo2 in regulating osteoblastogenesis and bone formation in postnatal bone has not been explored. Here we show that limb-bud progenitor cells from Rspo2 knockout mice undergo reduced mineralization during osteoblastogenesis in vitro and have a corresponding alteration in their osteogenic gene expression profile. We also generated the first Rspo2 conditional knockout (Rspo2floxed) mouse and disrupted Rspo2 expression in osteoblast-lineage cells by crossing to the Osteocalcin-Cre mouse line (Ocn-Cre + Rspo2f/f). Ocn-Cre + Rspo2f/f male and female mice at 1, 3, and 6 months were examined. Ocn-Cre + Rspo2f/f mice are decreased in overall body size compared to their control littermates and have decreased bone mass. Histomorphometric analysis of 1-month-old mice revealed a similar number of osteoblasts and mineralizing surface per bone surface with a simultaneous decrease in mineral apposition and bone formation rates. Consistent with this observation, serum osteocalcin in 3-month-old Ocn-Cre + Rspo2f/f was reduced, and bone marrow-mesenchymal stem cells from Ocn-Cre + Rspo2f/f mice undergo less mineralization in vitro. Finally, gene expression analysis and immunohistochemistry of mature bone shows reduced beta-catenin signaling in Ocn-Cre + Rspo2f/f. Overall, RSPO2 reduces osteoblastogenesis and mineralization, leading to reduced bone mass. A loss of R-spondin-2 reduces osteoblastogenesis (production of osteoblasts, the cells from which bone develops) and mineralization, thereby leading to decreased bone mass in adults. R-spondin-2 is one of a family of four proteins that are expressed in the developing mouse limb as well as other tissues; each R-spondin family member exerts a different functional effect. R-spondins clearly influence several aspects of skeletal biology, but their specific roles—especially in postnatal bone—remained to be elucidated. A team headed by Kurt Hankenson at the University of Michigan Medical School investigated the role of R-spondin-2 in osteoblastogenesis, both in vitro and in vivo, using a mouse model. For the first time, the team was able to demonstrate that R-spondin-2 promotes osteoblastogenesis, bone development, and consequent bone mass growth in adult mice.
Collapse
Affiliation(s)
- M Noelle Knight
- 1Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Kannan Karuppaiah
- 2Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, USA
| | - Michele Lowe
- 2Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, USA
| | - Sarthak Mohanty
- 1Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Robert L Zondervan
- 2Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, USA
| | - Sheila Bell
- 3Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Jaimo Ahn
- 1Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Kurt D Hankenson
- 1Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,2Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
28
|
Monteagudo S, Lories RJ. Cushioning the cartilage: a canonical Wnt restricting matter. Nat Rev Rheumatol 2017; 13:670-681. [PMID: 29021569 DOI: 10.1038/nrrheum.2017.171] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wnt signalling pathways have key roles in joint development, homeostasis and disease, particularly in osteoarthritis. New data is starting to reveal the importance of tightly regulating canonical Wnt signalling pathway activation to maintain homeostasis and health in articular cartilage. In addition to the presence of different Wnt antagonists that limit pathway activation in articular cartilage, the reciprocal crosstalk between the canonical and non-canonical cascades and competitive antagonism between different Wnt ligands seem to be critical in restraining excessive Wnt pathway activation. Changes in transcriptional complex assembly upon Wnt pathway activation, epigenetic modulation of target gene transcription, in particular through histone modifications, and complex interactions between the Wnt signalling pathway and other signalling pathways, are also instrumental in adjusting Wnt signalling. In this Review, the cellular and molecular mechanisms involved in fine-tuning canonical Wnt signalling in the joint are updated, with a focus on the articular cartilage. The interventions for preventing or treating osteoarthritis are also discussed, which should aim to limit disease-associated excessive canonical Wnt activity to avoid joint damage.
Collapse
Affiliation(s)
- Silvia Monteagudo
- Laboratory for Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Rik J Lories
- Laboratory for Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| |
Collapse
|
29
|
Pridgeon MG, Grohar PJ, Steensma MR, Williams BO. Wnt Signaling in Ewing Sarcoma, Osteosarcoma, and Malignant Peripheral Nerve Sheath Tumors. Curr Osteoporos Rep 2017. [PMID: 28647886 DOI: 10.1007/s11914-017-0377-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Wnt signaling plays a central role in development and homeostasis, and its dysregulation is a common event in many types of human cancer. Here we explore in detail the contributions of Wnt signaling to the initiation and maintenance of three types of saroma: Ewing sarcoma, osteosarcoma, and malignant peripheral nerve sheath tumors. This review provides an overview of the Wnt signaling pathway and explores in detail the current knowledge about its role in the initiation or maintenance of three tumor types: Ewing sarcoma, osteosarcoma, and malignant peripheral nerve sheath tumors. RECENT FINDINGS Recent work has assessed the role(s) of Wnt signaling within these cell types. This review provides an overview of the mechanistic insights that have been gained from a number of recent studies to set the foundation for potential therapeutic applications. Wnt signaling has emerged as a potentially critical pathway in maintaining the growth of these types of tumors. Given the fact that many new inhibitors of the pathway have recently or will soon enter Phase 1 clinical trials, it is likely that assessment of their activity in these tumor types will occur in human patients.
Collapse
Affiliation(s)
- Matthew G Pridgeon
- Grand Rapids Medical Education Partners, Grand Rapids, MI, USA
- Spectrum Health Cancer Center, Spectrum Health System, Grand Rapids, MI, USA
- Helen De Vos Children's Hospital, Grand Rapids, MI, USA
| | - Patrick J Grohar
- Spectrum Health Cancer Center, Spectrum Health System, Grand Rapids, MI, USA
- Helen De Vos Children's Hospital, Grand Rapids, MI, USA
- Department of Pediatrics, Michigan State University, Grand Rapids, MI, USA
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Matthew R Steensma
- Spectrum Health Cancer Center, Spectrum Health System, Grand Rapids, MI, USA
- Helen De Vos Children's Hospital, Grand Rapids, MI, USA
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
- Department of Surgery, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Bart O Williams
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA.
| |
Collapse
|
30
|
Dong X, Liao W, Zhang L, Tu X, Hu J, Chen T, Dai X, Xiong Y, Liang W, Ding C, Liu R, Dai J, Wang O, Lu L, Lu X. RSPO2 suppresses colorectal cancer metastasis by counteracting the Wnt5a/Fzd7-driven noncanonical Wnt pathway. Cancer Lett 2017; 402:153-165. [PMID: 28600110 DOI: 10.1016/j.canlet.2017.05.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/18/2017] [Accepted: 05/26/2017] [Indexed: 01/01/2023]
Abstract
R-spondins play critical roles in development, stem cell survival, and tumorigenicity by modulating Wnt/β-catenin signaling; however, the role of R-spondins in noncanonical Wnt signaling regulation remains largely unknown. We demonstrate here that R-spondin 2 (RSPO2) has an inhibitory effect on colorectal cancer (CRC) cell migration, invasion, and metastasis. Reduced RSPO2 expression was associated with tumor metastasis and poor survival in CRC patients. The metastasis-suppressive activity of RSPO2 was independent of the Wnt/β-catenin signaling pathway but dependent on the Fzd7-mediated noncanonical Wnt signaling pathway. The physical interaction of RSPO2 and Fzd7 increased the degradation of cell surface Fzd7 via ZNRF3-mediated ubiquitination, which led to the suppression of the downstream PKC/ERK signaling cascade. In late-stage metastatic cancer, Wnt5a promoted CRC cell migration by preventing degradation of Fzd7, and RSPO2 antagonized Wnt5a-driven noncanonical Wnt signaling activation and tumor cell migration by blocking the binding of Wnt5a to the Fzd7 receptor. Our study reveals a novel RSPO2/Wnt5a-competing noncanonical Wnt signaling mechanism that regulates cellular migration and invasion, and our data suggest that secreted RSPO2 protein could serve as a potential therapy for Wnt5a/Fzd7-driven aggressive CRC tumors.
Collapse
Affiliation(s)
- Xiaoming Dong
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wanqin Liao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Li Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xi Tu
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province, Zhejiang, 317000, China
| | - Jin Hu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Tianke Chen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaowei Dai
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Xiong
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Weicheng Liang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Chaodong Ding
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Rui Liu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Juji Dai
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ouchen Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Liting Lu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xincheng Lu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
31
|
Evidence of the Role of R-Spondin 1 and Its Receptor Lgr4 in the Transmission of Mechanical Stimuli to Biological Signals for Bone Formation. Int J Mol Sci 2017; 18:ijms18030564. [PMID: 28272338 PMCID: PMC5372580 DOI: 10.3390/ijms18030564] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 01/28/2023] Open
Abstract
The bone can adjust its mass and architecture to mechanical stimuli via a series of molecular cascades, which have been not yet fully elucidated. Emerging evidence indicated that R-spondins (Rspos), a family of secreted agonists of the Wnt/β-catenin signaling pathway, had important roles in osteoblastic differentiation and bone formation. However, the role of Rspo proteins in mechanical loading-influenced bone metabolism has never been investigated. In this study, we found that Rspo1 was a mechanosensitive protein for bone formation. Continuous cyclic mechanical stretch (CMS) upregulated the expression of Rspo1 in mouse bone marrow mesenchymal stem cells (BMSCs), while the expression of Rspo1 in BMSCs in vivo was downregulated in the bones of a mechanical unloading mouse model (tail suspension (TS)). On the other hand, Rspo1 could promote osteogenesis of BMSCs under CMS through activating the Wnt/β-catenin signaling pathway and could rescue the bone loss induced by mechanical unloading in the TS mice. Specifically, our results suggested that Rspo1 and its receptor of leucine-rich repeat containing G-protein-coupled receptor 4 (Lgr4) should be a novel molecular signal in the transmission of mechanical stimuli to biological signal in the bone, and this signal should be in the upstream of Wnt/β-catenin signaling for bone formation. Rspo1/Lgr4 could be a new potential target for the prevention and treatment of disuse osteoporosis in the future.
Collapse
|
32
|
Lu ZH, Kaliberov S, Sohn RE, Kaliberova L, Du Y, Prior JL, Leib DJ, Chauchereau A, Sehn JK, Curiel DT, Arbeit JM. A new model of multi-visceral and bone metastatic prostate cancer with perivascular niche targeting by a novel endothelial specific adenoviral vector. Oncotarget 2017; 8:12272-12289. [PMID: 28103576 PMCID: PMC5355343 DOI: 10.18632/oncotarget.14699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022] Open
Abstract
While modern therapies for metastatic prostate cancer (PCa) have improved survival they are associated with an increasingly prevalent entity, aggressive variant PCa (AVPCa), lacking androgen receptor (AR) expression, enriched for cancer stem cells (CSCs), and evidencing epithelial-mesenchymal plasticity with a varying extent of neuroendocrine transdifferentiation. Parallel work revealed that endothelial cells (ECs) create a perivascular CSC niche mediated by juxtacrine and membrane tethered signaling. There is increasing interest in pharmacological metastatic niche targeting, however, targeted access has been impossible. Here, we discovered that the Gleason 7 derived, androgen receptor negative, IGR-CaP1 cell line possessed some but not all of the molecular features of AVPCa. Intracardiac injection into NOD/SCID/IL2Rg -/- (NSG) mice produced a completely penetrant bone, liver, adrenal, and brain metastatic phenotype; noninvasively and histologically detectable at 2 weeks, and necessitating sacrifice 4-5 weeks post injection. Bone metastases were osteoblastic, and osteolytic. IGR-CaP1 cells expressed the neuroendocrine marker synaptophysin, near equivalent levels of vimentin and e-cadherin, all of the EMT transcription factors, and activation of NOTCH and WNT pathways. In parallel, we created a new triple-targeted adenoviral vector containing a fiber knob RGD peptide, a hexon mutation, and an EC specific ROBO4 promoter (Ad.RGD.H5/3.ROBO4). This vector was expressed in metastatic microvessels tightly juxtaposed to IGR-CaP1 cells in bone and visceral niches. Thus, the combination of IGR-CaP1 cells and NSG mice produces a completely penetrant metastatic PCa model emulating end-stage human disease. In addition, the metastatic niche access provided by our novel Ad vector could be therapeutically leveraged for future disease control or cure.
Collapse
Affiliation(s)
- Zhi Hong Lu
- Urology Division and Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Sergey Kaliberov
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Biologic Therapeutics Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca E. Sohn
- Urology Division and Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Lyudmila Kaliberova
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Biologic Therapeutics Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Yingqiu Du
- Urology Division and Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Julie L. Prior
- Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Daniel J. Leib
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Anne Chauchereau
- Prostate Cancer Group, INSERM U981, Gustave Roussy, Villejuif, F-94805, France
| | - Jennifer K. Sehn
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Anatomic and Molecular Pathology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - David T. Curiel
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Biologic Therapeutics Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey M. Arbeit
- Urology Division and Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
33
|
Alowolodu O, Johnson G, Alashwal L, Addou I, Zhdanova IV, Uversky VN. Intrinsic disorder in spondins and some of their interacting partners. INTRINSICALLY DISORDERED PROTEINS 2016; 4:e1255295. [PMID: 28232900 DOI: 10.1080/21690707.2016.1255295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 10/22/2016] [Accepted: 10/27/2016] [Indexed: 12/28/2022]
Abstract
Spondins, which are proteins that inhibit and promote adherence of embryonic cells so as to aid axonal growth are part of the thrombospondin-1 family. Spondins function in several important biological processes, such as apoptosis, angiogenesis, etc. Spondins constitute a thrombospondin subfamily that includes F-spondin, a protein that interacts with Aβ precursor protein and inhibits its proteolytic processing; R-spondin, a 4-membered group of proteins that regulates Wnt pathway and have other functions, such as regulation of kidney proliferation, induction of epithelial proliferation, the tumor suppressant action; M-spondin that mediates mechanical linkage between the muscles and apodemes; and the SCO-spondin, a protein important for neuronal development. In this study, we investigated intrinsic disorder status of human spondins and their interacting partners, such as members of the LRP family, LGR family, Frizzled family, and several other binding partners in order to establish the existence and importance of disordered regions in spondins and their interacting partners by conducting a detailed analysis of their sequences, finding disordered regions, and establishing a correlation between their structure and biological functions.
Collapse
Affiliation(s)
- Oluwole Alowolodu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Gbemisola Johnson
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Lamis Alashwal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Iqbal Addou
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Irina V Zhdanova
- Department of Anatomy & Neurobiology, Boston University School of Medicine , Boston, MA, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; USF Health Byrd Alzheimer Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
34
|
The role of R-spondins and their receptors in bone metabolism. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:93-100. [DOI: 10.1016/j.pbiomolbio.2016.05.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/27/2016] [Accepted: 05/24/2016] [Indexed: 12/21/2022]
|
35
|
Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: Therapeutic targets, active agents, and therapeutic localization. Adv Drug Deliv Rev 2016; 97:56-68. [PMID: 26763408 DOI: 10.1016/j.addr.2015.12.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.
Collapse
|
36
|
Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 2014; 37:1-14. [PMID: 25064829 PMCID: PMC4379989 DOI: 10.1016/j.matbio.2014.07.005] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
The concept of a matricellular protein was first proposed by Paul Bornstein in the mid-1990s to account for the non-lethal phenotypes of mice with inactivated genes encoding thrombospondin-1, tenascin-C, or SPARC. It was also recognized that these extracellular matrix proteins were primarily counter or de-adhesive. This review reappraises the matricellular concept after nearly two decades of continuous investigation. The expanded matricellular family as well as the diverse and often unexpected functions, cellular location, and interacting partners/receptors of matricellular proteins are considered. Development of therapeutic strategies that target matricellular proteins are discussed in the context of pathology and regenerative medicine.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | | |
Collapse
|