1
|
Zhai Y, Zhou Z, Xing X, Nuzzle M, Zhang X. Differential bone and vessel type formation at superior and dura periosteum during cranial bone defect repair. Bone Res 2025; 13:8. [PMID: 39805832 PMCID: PMC11729862 DOI: 10.1038/s41413-024-00379-9] [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: 04/17/2024] [Revised: 08/14/2024] [Accepted: 10/09/2024] [Indexed: 01/16/2025] Open
Abstract
The cranial mesenchyme, originating from both neural crest and mesoderm, imparts remarkable regional specificity and complexity to postnatal calvarial tissue. While the distinct embryonic origins of the superior and dura periosteum of the cranial parietal bone have been described, the extent of their respective contributions to bone and vessel formation during adult bone defect repair remains superficially explored. Utilizing transgenic mouse models in conjunction with high-resolution multiphoton laser scanning microscopy (MPLSM), we have separately evaluated bone and vessel formation in the superior and dura periosteum before and after injury, as well as following intermittent treatment of recombinant peptide of human parathyroid hormone (rhPTH), Teriparatide. Our results show that new bone formation along the dura surface is three times greater than that along the superior periosteal surface following injury, regardless of Teriparatide treatment. Targeted deletion of PTH receptor PTH1R via SMA-CreER and Col 1a (2.3)-CreER results in selective reduction of bone formation, suggesting different progenitor cell pools in the adult superior and dura periosteum. Consistently, analyses of microvasculature show higher vessel density and better organized arterial-venous vessel network associated with a 10-fold more osteoblast clusters at dura periosteum as compared to superior periosteum. Intermittent rhPTH treatment further enhances the arterial vessel ratio at dura periosteum and type H vessel formation in cortical bone marrow space. Taken together, our study demonstrates a site-dependent coordinated osteogenic and angiogenic response, which is determined by regional osteogenic progenitor pool as well as the coupling blood vessel network at the site of cranial defect repair.
Collapse
Affiliation(s)
- Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Mark Nuzzle
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
2
|
Skubic C, Trček H, Nassib P, Kreft T, Walakira A, Pohar K, Petek S, Režen T, Ihan A, Rozman D. Knockouts of CYP51A1, DHCR24, or SC5D from cholesterol synthesis reveal pathways modulated by sterol intermediates. iScience 2024; 27:110651. [PMID: 39262789 PMCID: PMC11387598 DOI: 10.1016/j.isci.2024.110651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Sterols from cholesterol synthesis are crucial for cholesterol production, but also have individual roles difficult to assess in vivo due to essentiality of cholesterol. We developed HepG2 cell models with knockouts (KOs) for three enzymes of cholesterol synthesis, each accumulating specific sterols. Surprisingly, KOs of CYP51, DHCR24, and SC5D shared only 9% of differentially expressed genes. The most striking was the phenotype of CYP51 KO with highly elevated lanosterol and 24,25-dihydrolanosterol, significant increase in G2+M phase and enhanced cancer and cell cycle pathways. Comparisons with mouse liver Cyp51 KO data suggest 24,25-dihydrolanosterol activates similar cell proliferation pathways, possibly via elevated LEF1 and WNT/NFKB signaling. In contrast, SC5D and DHCR24 KO cells with elevated lathosterol or desmosterol proliferated slowly, with downregulated E2F, mitosis, and enriched HNF1A. These findings demonstrate that increase of lanosterol and 24,25-dihydrolanosterol, but not other sterols, promotes cell proliferation in hepatocytes.
Collapse
Affiliation(s)
- Cene Skubic
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Hana Trček
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Petra Nassib
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Tinkara Kreft
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Andrew Walakira
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Katka Pohar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sara Petek
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Režen
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Liu H, Liu L, Rosen CJ. PTH and the Regulation of Mesenchymal Cells within the Bone Marrow Niche. Cells 2024; 13:406. [PMID: 38474370 PMCID: PMC10930661 DOI: 10.3390/cells13050406] [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: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Parathyroid hormone (PTH) plays a pivotal role in maintaining calcium homeostasis, largely by modulating bone remodeling processes. Its effects on bone are notably dependent on the duration and frequency of exposure. Specifically, PTH can initiate both bone formation and resorption, with the outcome being influenced by the manner of PTH administration: continuous or intermittent. In continuous administration, PTH tends to promote bone resorption, possibly by regulating certain genes within bone cells. Conversely, intermittent exposure generally favors bone formation, possibly through transient gene activation. PTH's role extends to various aspects of bone cell activity. It directly influences skeletal stem cells, osteoblastic lineage cells, osteocytes, and T cells, playing a critical role in bone generation. Simultaneously, it indirectly affects osteoclast precursor cells and osteoclasts, and has a direct impact on T cells, contributing to its role in bone resorption. Despite these insights, the intricate mechanisms through which PTH acts within the bone marrow niche are not entirely understood. This article reviews the dual roles of PTH-catabolic and anabolic-on bone cells, highlighting the cellular and molecular pathways involved in these processes. The complex interplay of these factors in bone remodeling underscores the need for further investigation to fully comprehend PTH's multifaceted influence on bone health.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Linyi Liu
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Clifford J. Rosen
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| |
Collapse
|
4
|
Lee SY, Fontana F, Sugatani T, Portales Castillo I, Leanza G, Coler-Reilly A, Civitelli R. Connexin43 in mesenchymal lineage cells regulates body adiposity and energy metabolism in mice. JCI Insight 2024; 9:e170016. [PMID: 38349739 PMCID: PMC11063945 DOI: 10.1172/jci.insight.170016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Connexin43 (Cx43) is the most abundant gap junction protein present in the mesenchymal lineage. In mature adipocytes, Cx43 mediates white adipose tissue (WAT) beiging in response to cold exposure and maintains the mitochondrial integrity of brown adipose tissue (BAT). We found that genetic deletion of Gja1 (Cx43 gene) in cells that give rise to chondro-osteogenic and adipogenic precursors driven by the Dermo1/Twist2 promoter led to lower body adiposity and partial protection against the weight gain and metabolic syndrome induced by a high-fat diet (HFD) in both sexes. These protective effects were related to increased locomotion, fuel utilization, energy expenditure, nonshivering thermogenesis, and better glucose tolerance in conditionally Gja1-ablated mice. Accordingly, Gja1-mutant mice exhibited reduced adipocyte hypertrophy, partially preserved insulin sensitivity, increased BAT lipolysis, and decreased whitening under HFD. This metabolic phenotype was not reproduced with more restricted Gja1 ablation in differentiated adipocytes, suggesting that Cx43 in adipocyte progenitors or other targeted cells restrains energy expenditures and promotes fat accumulation. These results reveal what we believe is a hitherto unknown action of Cx43 in adiposity, and offer a promising new pharmacologic target for improving metabolic balance in diabetes and obesity.
Collapse
|
5
|
Lee SY, Fontana F, Sugatani T, Castillo IP, Leanza G, Coler-Reilly A, Civitelli R. Connexin43 in mesenchymal lineage cells regulates body adiposity and energy metabolism in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574415. [PMID: 38260624 PMCID: PMC10802316 DOI: 10.1101/2024.01.05.574415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Connexin43 (Cx43) is the most abundant gap junction protein present in the mesenchymal lineage. In mature adipocytes, Cx43 mediates white adipose tissue (WAT) "beiging" in response to cold exposure and maintains the mitochondrial integrity of brown adipose tissue (BAT). We found that genetic deletion of Gja1 (Cx43 gene) in cells that give rise to chondro-osteogenic and adipogenic precursors driven by the Dermo1/Twist2 promoter leads to lower body adiposity and partial protection against the weight gain and metabolic syndrome induced by a high fat diet (HFD) in both sexes. These protective effects from obesogenic diet are related to increased locomotion, fuel utilization, energy expenditure, non-shivering thermogenesis, and better glucose tolerance in conditionally Gja1 ablated mice. Accordingly, Gja1 mutant mice exhibit reduced adipocyte hypertrophy, partially preserved insulin sensitivity, increased BAT lipolysis and decreased whitening under HFD. This metabolic phenotype is not reproduced with more restricted Gja1 ablation in differentiated adipocytes, suggesting that Cx43 has a hitherto unknown function in adipocyte progenitors or other targeted cells, resulting in restrained energy expenditures and fat accumulation. These results disclose an hitherto unknown action of Cx43 in adiposity, and offer a promising new pharmacologic target for improving metabolic balance in diabetes and obesity.
Collapse
Affiliation(s)
- Seung-Yon Lee
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Francesca Fontana
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Toshifumi Sugatani
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Ignacio Portales Castillo
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Giulia Leanza
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Ariella Coler-Reilly
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| | - Roberto Civitelli
- Department of Medicine, Division of Bone and Mineral Diseases; Musculoskeletal Research Center; Washington University School of Medicine, St. Louis, MO. USA
| |
Collapse
|
6
|
Kantaputra P, Jatooratthawichot P, Jintakanon K, Intachai W, Pradermdutsadeeporn P, Adisornkanj P, Tongsima S, Ngamphiw C, Olsen B, Tucker AS, Cairns JRK. Mutations in LRP6 highlight the role of WNT signalling in oral exostoses and dental anomalies. Arch Oral Biol 2022; 142:105514. [DOI: 10.1016/j.archoralbio.2022.105514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 07/28/2022] [Indexed: 02/07/2023]
|
7
|
St-Arnaud R, Pellicelli M, Ismail M, Arabian A, Jafarov T, Zhou CJ. NACA and LRP6 Are Part of a Common Genetic Pathway Necessary for Full Anabolic Response to Intermittent PTH. Int J Mol Sci 2022; 23:ijms23020940. [PMID: 35055125 PMCID: PMC8780913 DOI: 10.3390/ijms23020940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/28/2022] Open
Abstract
PTH induces phosphorylation of the transcriptional coregulator NACA on serine 99 through Gαs and PKA. This leads to nuclear translocation of NACA and expression of the target gene Lrp6, encoding a coreceptor of the PTH receptor (PTH1R) necessary for full anabolic response to intermittent PTH (iPTH) treatment. We hypothesized that maintaining enough functional PTH1R/LRP6 coreceptor complexes at the plasma membrane through NACA-dependent Lrp6 transcription is important to ensure maximal response to iPTH. To test this model, we generated compound heterozygous mice in which one allele each of Naca and Lrp6 is inactivated in osteoblasts and osteocytes, using a knock-in strain with a Naca99 Ser-to-Ala mutation and an Lrp6 floxed strain (test genotype: Naca99S/A; Lrp6+/fl;OCN-Cre). Four-month-old females were injected with vehicle or 100 μg/kg PTH(1-34) once daily, 5 days a week for 4 weeks. Control mice showed significant increases in vertebral trabecular bone mass and biomechanical properties that were abolished in compound heterozygotes. Lrp6 expression was reduced in compound heterozygotes vs. controls. The iPTH treatment increased Alpl and Col1a1 mRNA levels in the control but not in the test group. These results confirm that NACA and LRP6 form part of a common genetic pathway that is necessary for the full anabolic effect of iPTH.
Collapse
Affiliation(s)
- René St-Arnaud
- Research Centre, Shriners Hospital for Children—Canada, Montreal, QC H4A 0A9, Canada; (M.P.); (M.I.); (A.A.); (T.J.)
- Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 1A4, Canada
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 1A1, Canada
- Correspondence: ; Tel.: +1-514-282-7155; Fax: +1-514-842-5581
| | - Martin Pellicelli
- Research Centre, Shriners Hospital for Children—Canada, Montreal, QC H4A 0A9, Canada; (M.P.); (M.I.); (A.A.); (T.J.)
| | - Mahmoud Ismail
- Research Centre, Shriners Hospital for Children—Canada, Montreal, QC H4A 0A9, Canada; (M.P.); (M.I.); (A.A.); (T.J.)
| | - Alice Arabian
- Research Centre, Shriners Hospital for Children—Canada, Montreal, QC H4A 0A9, Canada; (M.P.); (M.I.); (A.A.); (T.J.)
| | - Toghrul Jafarov
- Research Centre, Shriners Hospital for Children—Canada, Montreal, QC H4A 0A9, Canada; (M.P.); (M.I.); (A.A.); (T.J.)
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA;
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children—Northern California, Sacramento, CA 95817, USA
| |
Collapse
|
8
|
Zweifler LE, Koh AJ, Daignault-Newton S, McCauley LK. Anabolic actions of PTH in murine models: two decades of insights. J Bone Miner Res 2021; 36:1979-1998. [PMID: 34101904 PMCID: PMC8596798 DOI: 10.1002/jbmr.4389] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 01/19/2023]
Abstract
Parathyroid hormone (PTH) is produced by the parathyroid glands in response to low serum calcium concentrations where it targets bones, kidneys, and indirectly, intestines. The N-terminus of PTH has been investigated for decades for its ability to stimulate bone formation when administered intermittently (iPTH) and is used clinically as an effective anabolic agent for the treatment of osteoporosis. Despite great interest in iPTH and its clinical use, the mechanisms of PTH action remain complicated and not fully defined. More than 70 gene targets in more than 90 murine models have been utilized to better understand PTH anabolic actions. Because murine studies utilized wild-type mice as positive controls, a variety of variables were analyzed to better understand the optimal conditions under which iPTH functions. The greatest responses to iPTH were in male mice, with treatment starting later than 12 weeks of age, a treatment duration lasting 5-6 weeks, and a PTH dose of 30-60 μg/kg/day. This comprehensive study also evaluated these genetic models relative to the bone formative actions with a primary focus on the trabecular compartment revealing trends in critical genes and gene families relevant for PTH anabolic actions. The summation of these data revealed the gene deletions with the greatest increase in trabecular bone volume in response to iPTH. These included PTH and 1-α-hydroxylase (Pth;1α(OH)ase, 62-fold), amphiregulin (Areg, 15.8-fold), and PTH related protein (Pthrp, 10.2-fold). The deletions with the greatest inhibition of the anabolic response include deletions of: proteoglycan 4 (Prg4, -9.7-fold), low-density lipoprotein receptor-related protein 6 (Lrp6, 1.3-fold), and low-density lipoprotein receptor-related protein 5 (Lrp5, -1.0-fold). Anabolic actions of iPTH were broadly affected via multiple and diverse genes. This data provides critical insight for future research and development, as well as application to human therapeutics. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Laura E Zweifler
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | | | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Pathology, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Bover J, Arana C, Ureña P, Torres A, Martín-Malo A, Fayos L, Coll V, Lloret MJ, Ochoa J, Almadén Y, Guirado L, Rodríguez M. Hyporesponsiveness or resistance to the action of parathyroid hormone in chronic kidney disease. Nefrologia 2021; 41:514-528. [PMID: 36165134 DOI: 10.1016/j.nefroe.2021.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/20/2020] [Indexed: 06/16/2023] Open
Abstract
Secondary hyperparathyroidism (SHPT) is an integral component of the chronic kidney disease-mineral and bone disorder (CKD-MBD). Many factors have been associated with the development and progression of SHPT but the presence of skeletal or calcemic resistance to the action of PTH in CKD has often gone unnoticed. The term hyporesponsiveness to PTH is currently preferred and, in this chapter, we will not only review the scientific timeline but also some of the molecular mechanisms behind. Moreover, the presence of resistance to the biological action of PTH is not unique in CKD since resistance to other hormones has also been described ("uremia as a receptor disease"). This hyporesponsiveness carries out important clinical implications since it explains, at least partially, not only the progressive nature of the pathogenesis of CKD-related PTH hypersecretion and parathyroid hyperplasia but also the increasing prevalence of adynamic bone disease in the CKD population. Therefore, we underline the importance of PTH control in all CKD stages, but not aiming to completely normalize PTH levels since a certain degree of SHPT may represent an adaptive clinical response. Future studies at the molecular level, i.e. on uremia or the recent description of the calcium-sensing receptor as a phosphate sensor, may become of great value beyond their significance to explain just the hyporesponsiveness to PTH in CKD.
Collapse
Affiliation(s)
- Jordi Bover
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain.
| | - Carolt Arana
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - Pablo Ureña
- AURA Nord Saint Ouen y Departamento de Fisiología Renal, Hospital Necker, Universidad de París Descartes, Paris, France
| | - Armando Torres
- Servicio de Nefrología, Hospital Universitario de Canarias, REDinREN, Universidad de La Laguna, Tenerife, Spain
| | - Alejandro Martín-Malo
- Unidad de Gestión Clinica Nefrología, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain; Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Leonor Fayos
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - Verónica Coll
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - María Jesús Lloret
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - Jackson Ochoa
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - Yolanda Almadén
- Unidad de Gestión Clínica Medicina Interna, Lipid and Atherosclerosis Unit, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluis Guirado
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - Mariano Rodríguez
- Unidad de Gestión Clinica Nefrología, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain; Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
El Khouri E, Ghoumid J, Haye D, Giuliano F, Drevillon L, Briand-Suleau A, De La Grange P, Nau V, Gaillon T, Bienvenu T, Jacquemin-Sablon H, Goossens M, Amselem S, Giurgea I. Wnt/β-catenin pathway and cell adhesion deregulation in CSDE1-related intellectual disability and autism spectrum disorders. Mol Psychiatry 2021; 26:3572-3585. [PMID: 33867523 DOI: 10.1038/s41380-021-01072-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 12/26/2022]
Abstract
Among the genetic factors playing a key role in the etiology of intellectual disabilities (IDs) and autism spectrum disorders (ASDs), several encode RNA-binding proteins (RBPs). In this study, we deciphered the molecular and cellular bases of ID-ASD in a patient followed from birth to the age of 21, in whom we identified a de novo CSDE1 (Cold Shock Domain-containing E1) nonsense variation. CSDE1 encodes an RBP that regulates multiple cellular pathways by monitoring the translation and abundance of target transcripts. Analyses performed on the patient's primary fibroblasts showed that the identified CSDE1 variation leads to haploinsufficiency. We identified through RNA-seq assays the Wnt/β-catenin signaling and cellular adhesion as two major deregulated pathways. These results were further confirmed by functional studies involving Wnt-specific luciferase and substrate adhesion assays. Additional data support a disease model involving APC Down-Regulated-1 (APCDD1) and cadherin-2 (CDH2), two components of the Wnt/β-catenin pathway, CDH2 being also pivotal for cellular adhesion. Our study, which relies on both the deep phenotyping and long-term follow-up of a patient with CSDE1 haploinsufficiency and on ex vivo studies, sheds new light on the CSDE1-dependent deregulated pathways in ID-ASD.
Collapse
Affiliation(s)
- E El Khouri
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - J Ghoumid
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille, France
| | - D Haye
- Service de Génétique Médicale Centre, Hospitalo-Universitaire de Nice, Nice, France
| | - F Giuliano
- Service de Génétique Médicale Centre, Hospitalo-Universitaire de Nice, Nice, France
| | - L Drevillon
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,CHU Caen Normandie, Caen, France
| | - A Briand-Suleau
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,Service de Génétique et Biologie Moléculaires, Hôpital Cochin, INSERM UMR1266 - Institute of Psychiatry and Neuroscience of Paris (IPNP) and University of Paris, Paris, France
| | | | - V Nau
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - T Gaillon
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France
| | - T Bienvenu
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, INSERM UMR1266 - Institute of Psychiatry and Neuroscience of Paris (IPNP) and University of Paris, Paris, France
| | - H Jacquemin-Sablon
- INSERM UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - M Goossens
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France
| | - S Amselem
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - I Giurgea
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France. .,Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.
| |
Collapse
|
11
|
Bover J, Arana C, Ureña P, Torres A, Martín-Malo A, Fayos L, Coll V, Lloret MJ, Ochoa J, Almadén Y, Guirado L, Rodríguez M. Hyporesponsiveness or resistance to the action of parathyroid hormone in chronic kidney disease. Nefrologia 2021. [PMID: 33985858 DOI: 10.1016/j.nefro.2020.12.019] [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] [Indexed: 12/12/2022] Open
Abstract
Secondary hyperparathyroidism (SHPT) is an integral component of the chronic kidney disease-mineral and bone disorder (CKD-MBD). Many factors have been associated with the development and progression of SHPT but the presence of skeletal or calcemic resistance to the action of PTH in CKD has often gone unnoticed. The term hyporesponsiveness to PTH is currently preferred and, in this chapter, we will not only review the scientific timeline but also some of the molecular mechanisms behind. Moreover, the presence of resistance to the biological action of PTH is not unique in CKD since resistance to other hormones has also been described ("uremia as a receptor disease"). This hyporesponsiveness carries out important clinical implications since it explains, at least partially, not only the progressive nature of the pathogenesis of CKD-related PTH hypersecretion and parathyroid hyperplasia but also the increasing prevalence of adynamic bone disease in the CKD population. Therefore, we underline the importance of PTH control in all CKD stages, but not aiming to completely normalize PTH levels since a certain degree of SHPT may represent an adaptive clinical response. Future studies at the molecular level, i.e. on uremia, or the recent description of the calcium-sensing receptor as a phosphate sensor, may become of great value beyond their significance to explain just the hyporesponsiveness to PTH in CKD.
Collapse
Affiliation(s)
- Jordi Bover
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España.
| | - Carolt Arana
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - Pablo Ureña
- AURA Nord Saint Ouen y Departamento de Fisiología Renal, Hospital Necker, Universidad de París Descartes, París, Francia
| | - Armando Torres
- Servicio de Nefrología, Hospital Universitario de Canarias, REDinREN, Universidad de La Laguna, Tenerife, España
| | - Alejandro Martín-Malo
- Unidad de Gestión Clínica Nefrología, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, España; Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Madrid, España
| | - Leonor Fayos
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - Verónica Coll
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - María Jesús Lloret
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - Jackson Ochoa
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - Yolanda Almadén
- Unidad de Gestión Clínica Medicina Interna, Lipid and Atherosclerosis Unit, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, España; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España
| | - Lluis Guirado
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, REDinREN, Barcelona, España
| | - Mariano Rodríguez
- Unidad de Gestión Clínica Nefrología, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, España; Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Madrid, España
| |
Collapse
|
12
|
N-cadherin in osteolineage cells modulates stromal support of tumor growth. J Bone Oncol 2021; 28:100356. [PMID: 33912383 PMCID: PMC8065282 DOI: 10.1016/j.jbo.2021.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/02/2022] Open
Abstract
N-cadherin in osteolineage, Osterix+ cells restrains extraskeletal tumor growth. Osterix+ cells are present in the stromal microenvironment of extraskeletal tumors. Osterix+ cells are present in normal tissues frequent sites of metastasis. N-cadherin modulates pro-tumorigenic signaling in tumor associated Osterix+ cells.
Tumor growth and metastases are dependent on interactions between cancer cells and the local environment. Expression of the cell–cell adhesion molecule N-cadherin (Ncad) is associated with highly aggressive cancers, and its expression by osteogenic cells has been proposed to provide a molecular “dock” for disseminated tumor cells to establish in pre-metastatic niches within the bone. To test this biologic model, we conditionally deleted the Ncad gene (Cdh2) in osteolineage cells using Osx-cre (cKO). Contrary to expectations, the metastatic breast cancer cell line PyMT-BO1 was able to form tumors in bone and to induce osteolysis in cKO as well as in control mice. Despite absence of Ncad, bone marrow stromal cells isolated from cKO mice were able to engage in direct cell–cell interactions with tumor cells expressing either N- or E-cadherin. However, subcutaneous PyMT-BO1 and B16F10 tumors grew larger in cKO relative to control littermates. Cell tracking experiments using the Ai9 reporter revealed the presence of Osx+ and Ncad+ cells in the stroma of extra-skeletal tumors and in a small population of lung cells. Gene expression analysis by RNAseq of Osx+ cells isolated from extra-skeletal tumors revealed alterations of pro-tumorigenic signaling pathways in cKO cells relative to control Osx+ cells. Thus, Ncad in Osx+ cells is not necessary for the establishment of bone metastases, but in extra-skeletal tumors it regulates pro-tumorigenic support by the microenvironment.
Collapse
|
13
|
Le PT, Liu H, Alabdulaaly L, Vegting Y, Calle IL, Gori F, Lanske B, Baron R, Rosen CJ. The role of Zfp467 in mediating the pro-osteogenic and anti-adipogenic effects on bone and bone marrow niche. Bone 2021; 144:115832. [PMID: 33359894 PMCID: PMC8175945 DOI: 10.1016/j.bone.2020.115832] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022]
Abstract
Conditional deletion of the PTH receptor (Pth1r) in mesenchymal progenitors reduces osteoblast differentiation and bone mass while enhancing adipogenesis and bone marrow adipose tissue. Mechanistically, PTH suppresses the expression of Zfp467, a pro-adipogenic zinc finger transcription factor. Consequently, Pth1r deficiency in mesenchymal progenitors leads to increased Zfp467 expression. Based on these observations, we hypothesized that genetic loss of Zfp467 would lead to a shift in marrow progenitor cell fate towards osteogenesis and increased bone mass. To test this hypothesis, we generated Zfp467-/- mice. Zfp467-/- mice (-/-) were significantly smaller than Zfp467+/+ mice (+/+). μCT showed significantly higher trabecular bone and cortical bone area in -/- vs. +/+, and histomorphometry showed higher structural and dynamic formation parameters in -/- mice vs. +/+. Femoral gene expression including Alpl, Sp7, and Acp5 were increased in -/-mice, whereas Adiponectin, Cebpa, Lepr, and Ppraγ mRNA were lower in -/- mice. Similarly, Fabp4 and Lep in the inguinal depot were also decreased in -/- mice. Moreover, marrow adipocyte numbers were reduced in -/- vs +/+ mice (p<0.007). In vitro, COBs and BMSCs-/- showed more positive ALP and Alizarin Red staining and a decrease in ORO droplets. Pth1r mRNA and protein levels were increased in COBs and BMSCs from -/- mice vs +/+ (p<0.02 for each parameter, -/- vs. +/+). -/- cells also exhibited enhanced endogenous levels of cAMP vs. control cells. Moreover, in an ovariectomy (OVX) mouse model, Zfp467-/- mice had significantly lower fat mass but similar bone mass compared to OVX +/+ mice. In contrast, in a high fat diet (HFD) mouse model, in addition to reduced adipocyte volume and adipogenesis related gene expression in both peripheral and bone marrow fat tissue, greater osteoblast number and higher osteogenesis related gene expression were also observed in -/- HFD mice vs. +/+ HFD mice. Taken together, these results demonstrate that ZFP467 negatively influences skeletal homeostasis and favors adipogenesis. Global deletion of Zfp467 increases PTHR1, cAMP and bone turnover, hence its repression is a component of PTH signaling and its regulation. These data support a critical role for Zfp467 in early lineage allocation and provide a novel potential mechanism by which PTH acts in an anabolic manner on the bone remodeling unit.
Collapse
Affiliation(s)
- Phuong T Le
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA
| | - Hanghang Liu
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA
| | - Lama Alabdulaaly
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Yosta Vegting
- University of Amsterdam, Amsterdam UMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
| | - Isabella L Calle
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA; Graduate Medical Sciences, Boston University School of Medicine, Boston, MA 02118, USA
| | - Francesca Gori
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Beate Lanske
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roland Baron
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Boston, MA 02115, USA; Harvard Medical School, Department of Medicine and Endocrine Unit, Massachusetts General Hospital, Boston, 02115, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA.
| |
Collapse
|
14
|
Hrdlicka HC, Pereira RC, Shin B, Yee SP, Deymier AC, Lee SK, Delany AM. Inhibition of miR-29-3p isoforms via tough decoy suppresses osteoblast function in homeostasis but promotes intermittent parathyroid hormone-induced bone anabolism. Bone 2021; 143:115779. [PMID: 33253931 PMCID: PMC7770763 DOI: 10.1016/j.bone.2020.115779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/07/2023]
Abstract
miRNAs play a vital role in post-transcriptional regulation of gene expression in osteoblasts and osteoclasts, and the miR-29 family is expressed in both lineages. Using mice globally expressing a miR-29-3p tough decoy, we demonstrated a modest 30-60% decrease all three miR-29-3p isoforms: miR-29a, miR-29b, and miR-29c. While the miR-29-3p decoy did not impact osteoclast number or function, the tough decoy decreased bone formation in growing mice, which led to decreased trabecular bone volume in mature animals. These data support previous in vitro studies suggesting that miR-29-3p is a positive regulator of osteoblast differentiation. In contrast, when mice were treated with intermittent parathyroid hormone (PTH1-34), inhibition of miR-29-3p augmented the effect of PTH on cortical bone anabolism, increased bone formation rate and osteoblast surface, and increased levels of Ctnnb1/βcatenin mRNA, which is a miR-29 target. These findings highlight differences in the mechanisms controlling basal level bone formation and bone formation induced by intermittent PTH. Overall, the global miR-29-3p tough decoy model represents a modest loss-of-function, which could be a relevant tool for assessing the possible impact of systemically administered miR-29-3p inhibitors. Our studies provide a potential rationale for co-administration of PTH1-34 and miR-29-3p inhibitors, to boost bone formation in severely affected osteoporosis patients, particularly in the cortical compartment.
Collapse
Affiliation(s)
- Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health Center, Farmington, CT, United States of America
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at University of California, Los Angeles, United States of America
| | - Bongjin Shin
- Center on Aging, UConn Health Center, Farmington, CT, United States of America
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, UConn Health Center, Farmington, CT, United States of America
| | - Alix C Deymier
- Institute of Material Sciences, UConn Health Center, Farmington, CT, United States of America
| | - Sun-Kyeong Lee
- Center on Aging, UConn Health Center, Farmington, CT, United States of America.
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health Center, Farmington, CT, United States of America.
| |
Collapse
|
15
|
Zhang C, Li T, Zhou C, Huang L, Li Y, Wang H, Duan P, Zou S, Mei L. Parathyroid hormone increases alveolar bone homoeostasis during orthodontic tooth movement in rats with periodontitis via crosstalk between STAT3 and β-catenin. Int J Oral Sci 2020; 12:38. [PMID: 33380723 PMCID: PMC7773736 DOI: 10.1038/s41368-020-00104-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/05/2023] Open
Abstract
Periodontitis patients are at risk of alveolar bone loss during orthodontic treatment. The aim of this study was to investigate whether intermittent parathyroid hormone (1–34) treatment (iPTH) could reduce alveolar bone loss during orthodontic tooth movement (OTM) in individuals with periodontitis and the underlying mechanism. A rat model of OTM in the context of periodontitis was established and alveolar bone loss was observed. The control, iPTH and iPTH + stattic groups received injections of vehicle, PTH and vehicle, or PTH and the signal transducer and activator of transcription 3 (STAT3) inhibitor stattic, respectively. iPTH prevented alveolar bone loss by enhancing osteogenesis and suppressing bone resorption in the alveolar bone during OTM in rats with periodontitis. This effect of iPTH was along with STAT3 activation and reduced by a local injection of stattic. iPTH promoted osteoblastic differentiation and might further regulate the Wnt/β-catenin pathway in a STAT3-dependent manner. The findings of this study suggest that iPTH might reduce alveolar bone loss during OTM in rats with periodontitis through STAT3/β-catenin crosstalk.
Collapse
Affiliation(s)
- Cheng Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tiancheng Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuyu Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Han Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peipei Duan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Li Mei
- Discipline of Orthodontics, Faculty of Dentistry, Department of Oral Sciences, Sir John Walsh Research Institute, University of Otago, Otago, New Zealand
| |
Collapse
|
16
|
Kapania EM, Reif TJ, Tsumura A, Eby JM, Callaci JJ. Alcohol-induced Wnt signaling inhibition during bone fracture healing is normalized by intermittent parathyroid hormone treatment. Animal Model Exp Med 2020; 3:200-207. [PMID: 32613179 PMCID: PMC7323703 DOI: 10.1002/ame2.12116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 11/14/2022] Open
Abstract
Nearly half of orthopaedic trauma patients are intoxicated at the time of injury, and excess alcohol consumption increases the risk for fracture nonunion. Previous studies show alcohol disrupts fracture associated Wnt signaling required for normal bone fracture repair. Intermittent parathyroid hormone (PTH) promotes bone growth through canonical Wnt signaling, however, no studies have investigated the effect of PTH on alcohol-inhibited bone fracture repair. Male C57BL/6 mice received two-3 day alcohol binges separated by 4 days before receiving a mid-shaft tibia fracture. Postoperatively, mice received PTH daily until euthanasia. Wnt/β-catenin signaling was analyzed at 9 days post-fracture. As previously observed, acute alcohol exposure resulted in a >2-fold decrease in total and the active form of β-catenin and a 2-fold increase in inactive β-catenin within the fracture callus. Intermittent PTH abrogated the effect of alcohol on β-catenin within the fracture callus. Upstream of β-catenin, alcohol-treated animals had a 2-fold decrease in total LRP6, the Wnt co-receptor, which was restored with PTH treatment. Alcohol nor PTH had any significant effect on GSK-3β. These data show that intermittent PTH following a tibia fracture restores normal expression of Wnt signaling proteins within the fracture callus of alcohol-treated mice.
Collapse
Affiliation(s)
- Esha M. Kapania
- Internal Medicine‐Pediatric ResidentRush University Medical CenterChicagoILUSA
| | - Taylor J. Reif
- Limb Lengthening and Complex Reconstruction FellowshipHospital for Special SurgeryNew YorkNYUSA
| | - Aaron Tsumura
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
| | - Jonathan M. Eby
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
- Alcohol Research Program (ARP)Loyola University Chicago Stritch School of MedicineMaywoodILUSA
| | - John J. Callaci
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
- Alcohol Research Program (ARP)Loyola University Chicago Stritch School of MedicineMaywoodILUSA
| |
Collapse
|
17
|
Revollo L, Kading J, Jeong SY, Li J, Salazar V, Mbalaviele G, Civitelli R. N-cadherin Restrains PTH Activation of Lrp6/β-catenin Signaling and Osteoanabolic Action. J Bone Miner Res 2019; 34:2163-2165. [PMID: 31721318 DOI: 10.1002/jbmr.3845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 11/06/2022]
|
18
|
Shi W, Ma Z, Zhang G, Wang C, Jiao Z. Novel functions of the primary cilium in bone disease and cancer. Cytoskeleton (Hoboken) 2019; 76:233-242. [PMID: 31108028 DOI: 10.1002/cm.21529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 11/09/2022]
Abstract
The primary cilium, a sensory organelle that emanates from the cell surface of most mammalian cell types during growth arrest, has attracted the attention of many researchers over the past decade. Recently, a large number of new findings have assigned novel functions and roles to the primary cilium in signal transduction and related diseases, which has greatly augmented the importance of the cilium in human health and development. Here, we review emerging evidence supporting the primary cilium as a sensory organelle in signal transduction in microgravity, electromagnetic field sensing, chemosensation and tumorigenesis. We also present an overview of signal transduction crosstalk associated with the primary cilium in bone disease and cancer, including primary cilium-related Ca2+ signaling, parathyroid hormone signaling, cAMP signaling, BMP/Smad1/5/8 signaling and Wnt signaling. We anticipate that emerging discoveries about the function of the primary cilium will provide novel insight into the molecular mechanisms of stimulus sensation, signal transduction and pathogenesis.
Collapse
Affiliation(s)
- Wengui Shi
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Zhijian Ma
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Gengyuan Zhang
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Chen Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.,The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Zuoyi Jiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.,The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| |
Collapse
|
19
|
Oxidized phospholipids are ligands for LRP6. Bone Res 2018; 6:22. [PMID: 30038821 PMCID: PMC6050227 DOI: 10.1038/s41413-018-0023-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/10/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023] Open
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) is a co-receptor for Wnt signaling and can be recruited by multiple growth factors/hormones to their receptors facilitating intracellular signaling activation. The ligands that bind directly to LRP6 have not been identified. Here, we report that bioactive oxidized phospholipids (oxPLs) are native ligands of LRP6, but not the closely related LRP5. oxPLs are products of lipid oxidation involving in pathological conditions such as hyperlipidemia, atherosclerosis, and inflammation. We found that cell surface LRP6 in bone marrow mesenchymal stromal cells (MSCs) decreased rapidly in response to increased oxPLs in marrow microenvironment. LRP6 directly bound and mediated the uptake of oxPLs by MSCs. oxPL-LRP6 binding induced LRP6 endocytosis through a clathrin-mediated pathway, decreasing responses of MSCs to osteogenic factors and diminishing osteoblast differentiation ability. Thus, LRP6 functions as a receptor and molecular target of oxPLs for their adverse effect on MSCs, revealing a potential mechanism underlying atherosclerosis-associated bone loss.
Collapse
|
20
|
Liang B, Huang J, Xu J, Li X, Li J. Local delivery of a novel PTHrP via mesoporous bioactive glass scaffolds to improve bone regeneration in a rat posterolateral spinal fusion model. RSC Adv 2018; 8:12484-12493. [PMID: 35539368 PMCID: PMC9079365 DOI: 10.1039/c8ra00870a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/26/2018] [Indexed: 12/01/2022] Open
Abstract
With the development of tissue engineering, bone defects, such as fractured long bones or cavitary lesions, may be efficiently repaired and reconstructed using bone substitutes. However, high rates of fusion failure remain unavoidable in spinal fusion surgery owing to the lack of appropriate materials for bone regeneration under such challenging conditions. Parathyroid hormone (PTH), a major regulator of bone remodeling, exerts both anabolic and catabolic effects. In this study, we modified PTH(1-34) and designed and synthesized a novel PTH-related peptide, namely PTHrP-1. Further, we fabricated a local PTHrP delivery device from mesoporous bioactive glass (MBG) to address the need for a suitable material in spinal fusion surgery. Using MBG scaffolds as a control, the biological properties of PTHrP-MBG scaffolds were evaluated in terms of attachment, proliferation, and alkaline phosphatase activity, as well as osteogenic gene and angiogenic gene expression in co-cultured rat bone marrow mesenchymal stem cells (rBMSCs) in vitro. Furthermore, PTHrP-1-MBG scaffolds were tested in a rat posterolateral spinal fusion model. Our data showed that PTHrP-1-MBG scaffolds possessed good ability to facilitate attachment and stimulation of rBMSC proliferation and differentiation. Importantly, the in vivo results revealed that the PTHrP-1-MBG scaffolds facilitated faster new bone formation and a higher rate and quality of spinal fusion. Therefore, the results suggest that devices consisting of the present novel PTHrP and MBG possess wider potential applications in bone regeneration and should serve as a promising bone substitute for spinal fusion.
Collapse
Affiliation(s)
- Bo Liang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital 600 Yishan Road Shanghai 200233 PR China
| | - Jinghuan Huang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital 600 Yishan Road Shanghai 200233 PR China
| | - Jianguang Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital 600 Yishan Road Shanghai 200233 PR China
| | - Xiaolin Li
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital 600 Yishan Road Shanghai 200233 PR China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University Wuhan 430071 PR China
| |
Collapse
|
21
|
Pellicelli M, Hariri H, Miller JA, St-Arnaud R. Lrp6 is a target of the PTH-activated αNAC transcriptional coregulator. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:61-71. [PMID: 29413898 DOI: 10.1016/j.bbagrm.2018.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/20/2022]
Abstract
In the nucleus of differentiated osteoblasts, the alpha chain of nascent polypeptide associated complex (αNAC) interacts with cJUN transcription factors to regulate the expression of target genes, including Osteocalcin (Bglap2). PTH induces the phosphorylation of αNAC on serine 99 through a Gαs-PKA dependent pathway. This leads to activation of αNAC and expression of Bglap2. To identify additional target genes regulated by PTH-activated αNAC, we performed ChIP-Seq against αNAC in PTH-treated MC3T3-E1 cells. This identified Low density lipoprotein receptor-Related Protein 6 (Lrp6) as a potential αNAC target. LRP6 acts as a co-receptor for the PTH receptor to allow optimal activation of PTH signaling. PTH increased Lrp6 mRNA levels in primary osteoblasts. Conventional quantitative ChIP confirmed the ChIP-Seq results. To assess whether αNAC plays a critical role in PTH-stimulated Lrp6 expression, we knocked-down Naca expression in MC3T3-E1 cells. Reduction of αNAC levels decreased basal expression of Lrp6 by 30% and blocked the stimulation of Lrp6 expression by PTH. We cloned the proximal mouse Lrp6 promoter (-2523/+120 bp) upstream of the luciferase reporter. Deletion and point mutations analysis in electrophoretic mobility shift assays and transient transfections identified a functional αNAC binding site centered around -343 bp. ChIP and ChIP-reChIP against JUND and αNAC showed that they cohabit on the proximal Lrp6 promoter. Luciferase assays confirmed that PTH-activated αNAC potentiated JUND-mediated Lrp6 transcription and Jund knockdown abolished this response. This study identified a novel αNAC target gene induced downstream of PTH signaling and represents the first characterization of the regulation of Lrp6 transcription in osteoblasts.
Collapse
Affiliation(s)
- Martin Pellicelli
- Research Centre, Shriners Hospitals for Children - Canada, H4A 0A9, Canada
| | - Hadla Hariri
- Research Centre, Shriners Hospitals for Children - Canada, H4A 0A9, Canada; Department of Human Genetics, McGill University, H3A 1A1, Canada
| | - Julie A Miller
- Research Centre, Shriners Hospitals for Children - Canada, H4A 0A9, Canada; Department of Human Genetics, McGill University, H3A 1A1, Canada
| | - René St-Arnaud
- Research Centre, Shriners Hospitals for Children - Canada, H4A 0A9, Canada; Department of Human Genetics, McGill University, H3A 1A1, Canada; Department of Surgery, McGill University, H3A 1A1, Canada; Department of Medicine, McGill University, H3A 1A1, Canada; Research Institute of the McGill University Health Centre, Montreal, Quebec H3H 2R9, Canada.
| |
Collapse
|
22
|
Marie PJ, Cohen-Solal M. The Expanding Life and Functions of Osteogenic Cells: From Simple Bone-Making Cells to Multifunctional Cells and Beyond. J Bone Miner Res 2018; 33:199-210. [PMID: 29206311 DOI: 10.1002/jbmr.3356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022]
Abstract
During the last three decades, important progress in bone cell biology and in human and mouse genetics led to major advances in our understanding of the life and functions of cells of the osteoblast lineage. Previously unrecognized sources of osteogenic cells have been identified. Novel cellular and molecular mechanisms controlling osteoblast differentiation and senescence have been determined. New mechanisms of communications between osteogenic cells, osteocytes, osteoclasts, and chondrocytes, as well as novel links between osteogenic cells and blood vessels have been identified. Additionally, cells of the osteoblast lineage were shown to be important components of the hematopoietic niche and to be implicated in hematologic dysfunctions and malignancy. Lastly, unexpected interactions were found between osteogenic cells and several soft tissues, including the central nervous system, gut, muscle, fat, and testis through the release of paracrine factors, making osteogenic cells multifunctional regulatory cells, in addition to their bone-making function. These discoveries considerably enlarged our vision of the life and functions of osteogenic cells, which may lead to the development of novel therapeutics with immediate applications in bone disorders. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Pierre J Marie
- Inserm UMR-1132, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Martine Cohen-Solal
- Inserm UMR-1132, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
23
|
Mansoori MN, Shukla P, Singh D. Combination of PTH (1-34) with anti-IL17 prevents bone loss by inhibiting IL-17/N-cadherin mediated disruption of PTHR1/LRP-6 interaction. Bone 2017; 105:226-236. [PMID: 28935557 DOI: 10.1016/j.bone.2017.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/17/2017] [Accepted: 09/17/2017] [Indexed: 12/18/2022]
Abstract
Combinations of anabolic and anti-resorptive agents have potential to improve bone density more than either agent alone. In this study, we determine the combining effect of anti-IL17 antibody and PTH (1-34) in mitigation of ovariectomy induced bone loss. Ovariectomized BALB/c female mice were treated with anti-IL17 and iPTH monotherapies and their combination. Combination of iPTH and anti-IL17 has synergistic effect in the restoration of skeletal and immune parameters compared to mono-therapies. Immunofluorescence analysis shows decreased expression of PTHR1 in iPTH+anti-IL17 treated bone sections. Our studies show that IL-17 up regulates N-cadherin which disrupts PTHR1/LRP-6 interaction thereby inhibiting wnt signaling and promoting bone loss. Our studies advocate use of iPTH and anti-IL17 combination therapy for post-menopausal osteoporosis.
Collapse
Affiliation(s)
- Mohd Nizam Mansoori
- Division of Endocrinology, Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India
| | - Priyanka Shukla
- Division of Endocrinology, Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India
| | - Divya Singh
- Division of Endocrinology, Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India.
| |
Collapse
|
24
|
Fontana F, Hickman-Brecks CL, Salazar VS, Revollo L, Abou-Ezzi G, Grimston SK, Jeong SY, Watkins M, Fortunato M, Alippe Y, Link DC, Mbalaviele G, Civitelli R. N-cadherin Regulation of Bone Growth and Homeostasis Is Osteolineage Stage-Specific. J Bone Miner Res 2017; 32:1332-1342. [PMID: 28240364 PMCID: PMC5466462 DOI: 10.1002/jbmr.3112] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/30/2017] [Accepted: 02/20/2017] [Indexed: 12/15/2022]
Abstract
N-cadherin inhibits osteogenic cell differentiation and canonical Wnt/β-catenin signaling in vitro. However, in vivo both conditional Cdh2 ablation and overexpression in osteoblasts lead to low bone mass. We tested the hypothesis that N-cadherin has different effects on osteolineage cells depending upon their differentiation stage. Embryonic conditional osteolineage Cdh2 deletion in mice results in defective growth, low bone mass, and reduced osteoprogenitor number. These abnormalities are prevented by delaying Cdh2 ablation until 1 month of age, thus targeting only committed and mature osteoblasts, suggesting they are the consequence of N-cadherin deficiency in osteoprogenitors. Indeed, diaphyseal trabecularization actually increases when Cdh2 is ablated postnatally. The sclerostin-insensitive Lrp5A214V mutant, associated with high bone mass, does not rescue the growth defect, but it overrides the low bone mass of embryonically Cdh2-deleted mice, suggesting N-cadherin interacts with Wnt signaling to control bone mass. Finally, bone accrual and β-catenin accumulation after administration of an anti-Dkk1 antibody are enhanced in N-cadherin-deficient mice. Thus, although lack of N-cadherin in embryonic and perinatal age is detrimental to bone growth and bone accrual, in adult mice loss of N-cadherin in osteolineage cells favors bone formation. Hence, N-cadherin inhibition may widen the therapeutic window of osteoanabolic agents. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Francesca Fontana
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Cynthia L Hickman-Brecks
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Valerie S Salazar
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Development Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Leila Revollo
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Development Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Grazia Abou-Ezzi
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Division of Oncology, Stem Cell Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan K Grimston
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sung Yeop Jeong
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Marcus Watkins
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Manuela Fortunato
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yael Alippe
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel C Link
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Division of Oncology, Stem Cell Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel Mbalaviele
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Roberto Civitelli
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
25
|
Yang Y, Lei H, Qiang YW, Wang B. Ixazomib enhances parathyroid hormone-induced β-catenin/T-cell factor signaling by dissociating β-catenin from the parathyroid hormone receptor. Mol Biol Cell 2017; 28:1792-1803. [PMID: 28495797 PMCID: PMC5491187 DOI: 10.1091/mbc.e17-02-0096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/15/2022] Open
Abstract
The proteasome inhibitor ixazomib (Izb) dissociates β-catenin from the PTH receptor to enhance PTH stimulation of β-catenin/TCF signaling through the cAMP/PKA signaling pathway. These findings provide a rationale for the use of Izb as an adjunct in the treatment of osteoporosis with PTH. The anabolic action of PTH in bone is mostly mediated by cAMP/PKA and Wnt-independent activation of β-catenin/T-cell factor (TCF) signaling. β-Catenin switches the PTH receptor (PTHR) signaling from cAMP/PKA to PLC/PKC activation by binding to the PTHR. Ixazomib (Izb) was recently approved as the first orally administered proteasome inhibitor for the treatment of multiple myeloma; it acts in part by inhibition of pathological bone destruction. Proteasome inhibitors were reported to stabilize β-catenin by the ubiquitin-proteasome pathway. However, how Izb affects PTHR activation to regulate β-catenin/TCF signaling is poorly understood. In the present study, using CRISPR/Cas9 genome-editing technology, we show that Izb reverses β-catenin–mediated PTHR signaling switch and enhances PTH-induced cAMP generation and cAMP response element–luciferase activity in osteoblasts. Izb increases active forms of β-catenin and promotes β-catenin translocation, thereby dissociating β-catenin from the PTHR at the plasma membrane. Furthermore, Izb facilitates PTH-stimulated GSK3β phosphorylation and β-catenin phosphorylation. Thus Izb enhances PTH stimulation of β-catenin/TCF signaling via cAMP-dependent activation, and this effect is due to its separating β-catenin from the PTHR. These findings provide evidence that Izb may be used to improve the therapeutic efficacy of PTH for the treatment of osteoporosis and other resorptive bone diseases.
Collapse
Affiliation(s)
- Yanmei Yang
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Hong Lei
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107.,College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Ya-Wei Qiang
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Bin Wang
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
26
|
|
27
|
Osagie-Clouard L, Sanghani A, Coathup M, Briggs T, Bostrom M, Blunn G. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017; 6:14-21. [PMID: 28062525 PMCID: PMC5227055 DOI: 10.1302/2046-3758.61.bjr-2016-0085.r1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Intermittently administered parathyroid hormone (PTH 1-34) has been shown to promote bone formation in both human and animal studies. The hormone and its analogues stimulate both bone formation and resorption, and as such at low doses are now in clinical use for the treatment of severe osteoporosis. By varying the duration of exposure, parathyroid hormone can modulate genes leading to increased bone formation within a so-called 'anabolic window'. The osteogenic mechanisms involved are multiple, affecting the stimulation of osteoprogenitor cells, osteoblasts, osteocytes and the stem cell niche, and ultimately leading to increased osteoblast activation, reduced osteoblast apoptosis, upregulation of Wnt/β-catenin signalling, increased stem cell mobilisation, and mediation of the RANKL/OPG pathway. Ongoing investigation into their effect on bone formation through 'coupled' and 'uncoupled' mechanisms further underlines the impact of intermittent PTH on both cortical and cancellous bone. Given the principally catabolic actions of continuous PTH, this article reviews the skeletal actions of intermittent PTH 1-34 and the mechanisms underlying its effect. CITE THIS ARTICLE L. Osagie-Clouard, A. Sanghani, M. Coathup, T. Briggs, M. Bostrom, G. Blunn. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017;6:14-21. DOI: 10.1302/2046-3758.61.BJR-2016-0085.R1.
Collapse
Affiliation(s)
- L Osagie-Clouard
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - A Sanghani
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Coathup
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - T Briggs
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Bostrom
- Hospital for Special Surgery, New York, New York, USA
| | - G Blunn
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| |
Collapse
|
28
|
Yang H, Dong J, Xiong W, Fang Z, Guan H, Li F. N-cadherin restrains PTH repressive effects on sclerostin/SOST by regulating LRP6-PTH1R interaction. Ann N Y Acad Sci 2016; 1385:41-52. [PMID: 27723935 DOI: 10.1111/nyas.13221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022]
Abstract
Sclerostin/SOST is a robust negative regulator of bone formation. Loss-of-function mutations of the sclerostin gene (SOST) cause sclerosteosis and Van Buchem disease characterized by bone overgrowth. Mediated by myocyte enhancer factor 2 (MEF2) transcription factors, parathyroid hormone (PTH) suppresses SOST expression through formation of complexes of parathyroid hormone-parathyroid hormone-related peptide receptor 1 (PTH1R) and lipoprotein receptor-related protein 6 (LRP6). N-cadherin has been shown to negatively regulate Wnt/β-catenin and PTH induced, protein kinase-dependent β-catenin signaling. Here, we investigated whether N-cadherin mediates the inhibitory effects of PTH on sclerostin/SOST. In vitro, overexpression of N-cadherin resulted in blunted PTH suppressive effects on sclerostin/SOST expression, as detected by immunoblot and qPCR analysis; PTH-induced downregulation of MEF2A, C, and D was impaired by N-cadherin; and N-cadherin reduced LRP6-PTHR1 interaction and endocytosis in response to PTH. In vivo, intermittent PTH (iPTH)-induced suppression of sclerostin/SOST was accentuated in Dmp1-cre; Cdh2f/f (Cdh2ΔDmp1 ) mice, compared with Cdh2f/f mice. Additionally, iPTH had greater bone anabolic effects in Cdh2ΔDmp1 mice compared to Cdh2f/f mice. These data indicate that N-cadherin negatively mediates PTH suppressive effects on sclerostin/SOST by regulating LRP6-PTHR1 interaction, ultimately influencing PTH anabolic effects on bone.
Collapse
Affiliation(s)
- Hailin Yang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China.,Department of Orthopaedics, People's Hospital of Jieshou City, Jieshou, Anhui, P.R. China
| | - Jinbo Dong
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Wei Xiong
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Zhong Fang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Hanfeng Guan
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Feng Li
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
29
|
Kedlaya R, Kang KS, Hong JM, Bettagere V, Lim KE, Horan D, Divieti-Pajevic P, Robling AG. Adult-Onset Deletion of β-Catenin in (10kb)Dmp1-Expressing Cells Prevents Intermittent PTH-Induced Bone Gain. Endocrinology 2016; 157:3047-57. [PMID: 27253995 PMCID: PMC4967118 DOI: 10.1210/en.2015-1587] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
β-Catenin (βcat) is a major downstream signaling node in canonical Wingless-related integration site (Wnt) signaling pathway, and its activity is crucial for canonical Wnt signal transduction. Wnt signaling has recently been implicated in the osteo-anabolic response to PTH, a potent calcium-regulating factor. We investigated whether βcat is essential for the anabolic action of intermittent PTH by generating male mice with adult-onset deletion of βcat in a subpopulation of bone cells (osteocytes and late-stage osteoblasts), treating them with an anabolic regimen of PTH, and measuring the skeletal responses. Male (10kb)Dmp1-CreERt2 transgenic mice that also harbored floxed loss-of-function βcat alleles (βcat(f/f)) were induced for Cre activity using tamoxifen, then injected daily with human PTH 1-34 (30 μg/kg) or vehicle for 5 weeks. Mice in which βcat was deleted showed either total lack of bone mineral density (BMD) gain, or BMD loss, and did not respond to PTH treatment. However, bone mass measurements in the trabecular compartment of the femur and spine revealed PTH-induced bone gain whether βcat was deleted or not. PTH-stimulated increases in periosteal and cancellous bone formation rates were not impaired by βcat deletion, but resorption markers and cortical porosity were significantly increased in induced mice, particularly induced mice treated with PTH. These results suggest that βcat is required for net-positive BMD effects of PTH therapy but that the anabolic effects per se of PTH treatment might not require osteocytic/osteoblastic βcat.
Collapse
Affiliation(s)
- Rajendra Kedlaya
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Kyung Shin Kang
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Jung Min Hong
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Vidya Bettagere
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Kyung-Eun Lim
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Daniel Horan
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Paola Divieti-Pajevic
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| | - Alexander G Robling
- Department of Anatomy and Cell Biology (R.K., K.S.K., J.M.H., V.B., K.-E.L., D.H., A.G.R.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Molecular and Cell Biology (P.D.-P.), Boston University School of Dental Medicine, Boston, Massachusetts 02215; Department of Biomedical Engineering (A.G.R.), Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202; and Richard L. Roudebush Veterans Affairs Medical Center (A.G.R.), Indianapolis, Indiana 46202
| |
Collapse
|
30
|
Estus TL, Choudhary S, Pilbeam CC. Prostaglandin-mediated inhibition of PTH-stimulated β-catenin signaling in osteoblasts by bone marrow macrophages. Bone 2016; 85:123-30. [PMID: 26851123 PMCID: PMC4835216 DOI: 10.1016/j.bone.2016.01.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 11/29/2022]
Abstract
Bone marrow macrophages (BMMs), in the presence of cyclooxygenase-2 (Cox2) produced PGE2, secrete an inhibitory factor in response to Rankl that blocks PTH-stimulated osteoblastic differentiation. This study was to determine if the inhibitory factor also blocks PTH-stimulated Wnt signaling. Primary calvarial osteoblasts (POBs) were co-cultured with conditioned medium (CM) from Rankl-treated wild type (WT) BMMs, which make the inhibitory factor, and Cox2 knockout (KO) BMMs, which do not. PTH induced cAMP production was blocked by WT CM but not by KO CM. In the presence of KO CM, PTH induced phosphorylation at β-catenin serine sites, ser552 and ser675, previously shown to be phosphorylated by protein kinase A (PKA). Phosphorylation was blocked by WT CM and by H89, a PKA inhibitor. PTH did not increase total β-catenin. PTH-stimulated transcription factor/lymphoid enhancer-binding factor response element activity in POBs was blocked by WT CM and by serum amyloid A (SAA), the human recombinant analog of murine Saa3, which has recently been shown to be the inhibitory factor. In POBs cultured with Cox2 KO CM, PTH increased expression of multiple genes associated with the anabolic actions of PTH and decreased expression of Wnt antagonists. This differential regulation of gene expression was not seen in POBs cultured with WT CM. These data highlight the ability of PTH to phosphorylate β-catenin directly via PKA and demonstrate the ability of a Cox2-dependent inhibitory factor, secreted by Rankl-stimulated BMMs, to abrogate PTH stimulated β-catenin signaling. Our results suggest that PTH can stimulate a novel negative feedback of its anabolic actions by stimulating Rankl and Cox2 expression.
Collapse
Affiliation(s)
- Thomas L Estus
- Department of Biomedical Engineering, University of Connecticut, Storrs, 263 Farmington Ave, Farmington, CT 06030, CT, United States; New England Musculoskeletal Institute, UConn Health, 263 Farmington Ave, Farmington, CT 06030, CT, United States.
| | - Shilpa Choudhary
- New England Musculoskeletal Institute, UConn Health, 263 Farmington Ave, Farmington, CT 06030, CT, United States; Department of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT 06030, CT, United States.
| | - Carol C Pilbeam
- Department of Biomedical Engineering, University of Connecticut, Storrs, 263 Farmington Ave, Farmington, CT 06030, CT, United States; New England Musculoskeletal Institute, UConn Health, 263 Farmington Ave, Farmington, CT 06030, CT, United States; Department of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT 06030, CT, United States.
| |
Collapse
|
31
|
Li C, Wang W, Xie L, Luo X, Cao X, Wan M. Lipoprotein receptor-related protein 6 is required for parathyroid hormone-induced Sost suppression. Ann N Y Acad Sci 2015; 1364:62-73. [PMID: 25847683 DOI: 10.1111/nyas.12750] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/02/2015] [Accepted: 02/19/2015] [Indexed: 02/05/2023]
Abstract
Parathyroid hormone (PTH) suppresses the expression of the bone formation inhibitor sclerostin (Sost) in osteocytes by inducing nuclear accumulation of histone deacetylases (HDACs) to inhibit the myocyte enhancer factor 2 (MEF2)-dependent Sost bone enhancer. Previous studies revealed that lipoprotein receptor-related protein 6 (LRP6) mediates the intracellular signaling activation and the anabolic bone effect of PTH. Here, we investigated whether LRP6 mediates the inhibitory effect of PTH on Sost using an osteoblast-specific Lrp6-knockout (LRP6-KO) mouse model. An increased level of Sost mRNA expression was detected in femur tissue from LRP6-KO mice, compared to wild-type littermates. The number of osteocytes expressing sclerostin protein was also increased in bone tissue of LRP6-KO littermates, indicating a negative regulatory role of LRP6 on Sost/sclerostin. In wild-type littermates, intermittent PTH treatment significantly suppressed Sost mRNA expression in bone and the number of sclerostin(+) osteocytes, while the effect of PTH was much less significant in LRP6-KO mice. Additionally, PTH-induced downregulation of MEF2C and 2D, as well as HDAC changes in osteocytes, were abrogated in LRP6-KO mice. These data indicate that LRP6 is required for PTH suppression of Sost expression.
Collapse
Affiliation(s)
- Changjun Li
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Institute of Endocrinology and Metabolism, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weishan Wang
- Department of Orthopaedics, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Liang Xie
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xianghang Luo
- Institute of Endocrinology and Metabolism, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Marie PJ. Osteoblast dysfunctions in bone diseases: from cellular and molecular mechanisms to therapeutic strategies. Cell Mol Life Sci 2015; 72:1347-61. [PMID: 25487608 PMCID: PMC11113967 DOI: 10.1007/s00018-014-1801-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/13/2014] [Accepted: 12/01/2014] [Indexed: 12/27/2022]
Abstract
Several metabolic, genetic and oncogenic bone diseases are characterized by defective or excessive bone formation. These abnormalities are caused by dysfunctions in the commitment, differentiation or survival of cells of the osteoblast lineage. During the recent years, significant advances have been made in our understanding of the cellular and molecular mechanisms underlying the osteoblast dysfunctions in osteoporosis, skeletal dysplasias and primary bone tumors. This led to suggest novel therapeutic approaches to correct these abnormalities such as the modulation of WNT signaling, the pharmacological modulation of proteasome-mediated protein degradation, the induction of osteoprogenitor cell differentiation, the repression of cancer cell proliferation and the manipulation of epigenetic mechanisms. This article reviews our current understanding of the major cellular and molecular mechanisms inducing osteoblastic cell abnormalities in age-related bone loss, genetic skeletal dysplasias and primary bone tumors, and discusses emerging therapeutic strategies to counteract the osteoblast abnormalities in these disorders of bone formation.
Collapse
Affiliation(s)
- Pierre J Marie
- INSERM UMR-1132, Hôpital Lariboisière, 2 rue Ambroise Paré, 75475, Paris Cedex 10, France,
| |
Collapse
|
33
|
Samuelov L, Sprecher E, Paus R. The role of P-cadherin in skin biology and skin pathology: lessons from the hair follicle. Cell Tissue Res 2015; 360:761-71. [PMID: 25707507 DOI: 10.1007/s00441-015-2114-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/18/2014] [Indexed: 12/21/2022]
Abstract
Adherens junctions (AJs) are one of the major intercellular junctions in various epithelia including the epidermis and the follicular epithelium. AJs connect the cell surface to the actin cytoskeleton and comprise classic transmembrane cadherins, such as P-cadherin, armadillo family proteins, and actin microfilaments. Loss-of-function mutations in CDH3, which encodes P-cadherin, result in two allelic autosomal recessive disorders: hypotrichosis with juvenile macular dystrophy (HJMD) and ectodermal dysplasia, ectrodactyly, and macular dystrophy (EEM) syndromes. Both syndromes feature sparse hair heralding progressive macular dystrophy. EEM syndrome is characterized in addition by ectodermal and limb defects. Recent studies have demonstrated that, together with its involvement in cell-cell adhesion, P-cadherin plays a crucial role in regulating cell signaling, malignant transformation, and other major intercellular processes. Here, we review the roles of P-cadherin in skin and hair biology, with emphasize on human hair growth, cycling and pigmentation.
Collapse
Affiliation(s)
- Liat Samuelov
- Department of Dermatology, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, Tel Aviv, 64239, Israel,
| | | | | |
Collapse
|