1
|
Kim JH, Kim K, Kim I, Seong S, Koh JT, Kim N. Stanniocalcin 1 and 1,25-dihydroxyvitamin D 3 cooperatively regulate bone mineralization by osteoblasts. Exp Mol Med 2024; 56:1991-2001. [PMID: 39218976 PMCID: PMC11447260 DOI: 10.1038/s12276-024-01302-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 09/04/2024] Open
Abstract
Stanniocalcin 1 (STC1) is a calcium- and phosphate-regulating hormone that is expressed in all tissues, including bone tissues, and is involved in calcium and phosphate homeostasis. Previously, STC1 expression was found to be increased by 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] administration in renal proximal tubular cells. In this study, we investigated whether STC1 directly regulates osteoblast differentiation or reciprocally controls the effects of 1,25(OH)2D3 on osteoblasts to contribute to bone homeostasis. We found that STC1 inhibited osteoblast differentiation in vitro and bone morphogenetic protein 2 (BMP2)-induced ectopic bone formation in vivo. Moreover, 1,25(OH)2D3 increased STC1 expression through direct binding to the Stc1 promoter of the vitamin D receptor (VDR). STC1 activated the 1,25(OH)2D3-VDR signaling pathway through the upregulation of VDR expression mediated by the inhibition of Akt phosphorylation in osteoblasts. STC1 further increased the effects of 1,25(OH)2D3 on receptor activator of nuclear factor-κB ligand (RANKL) secretion and inhibited osteoblast differentiation by exhibiting a positive correlation with 1,25(OH)2D3. The long-bone phenotype of transgenic mice overexpressing STC1 specifically in osteoblasts was not significantly different from that of wild-type mice. However, compared with that in the wild-type mice, 1,25(OH)2D3 administration significantly decreased bone mass in the STC1 transgenic mice. Collectively, these results suggest that STC1 negatively regulates osteoblast differentiation and bone formation; however, the inhibitory effect of STC1 on osteoblasts is transient and can be reversed under normal conditions. Nevertheless, the synergistic effect of STC1 and 1,25(OH)2D3 through 1,25(OH)2D3 administration may reduce bone mass by inhibiting osteoblast differentiation.
Collapse
Affiliation(s)
- Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
2
|
Khatun M, Modhukur V, Piltonen TT, Tapanainen JS, Salumets A. Stanniocalcin Protein Expression in Female Reproductive Organs: Literature Review and Public Cancer Database Analysis. Endocrinology 2024; 165:bqae110. [PMID: 39186548 PMCID: PMC11398916 DOI: 10.1210/endocr/bqae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/24/2024] [Indexed: 08/28/2024]
Abstract
Stanniocalcin (STC) 1 and 2 serve as antihyperglycemic polypeptide hormones with critical roles in regulating calcium and phosphate homeostasis. They additionally function as paracrine and/or autocrine factors involved in numerous physiological processes, including female reproduction. STC1 and STC2 contribute to the pathophysiology of several diseases, including female infertility- and pregnancy-associated conditions, and even tumorigenesis of reproductive organs. This comprehensive review highlights the dynamic expression patterns and potential dysregulation of STC1 and STC2, restricted to female fertility, and infertility- and pregnancy-associated diseases and conditions, such as endometriosis, polycystic ovary syndrome (PCOS), abnormal uterine bleeding, uterine polyps, and pregnancy complications, like impaired decidualization, preeclampsia, and preterm labor. Furthermore, the review elucidates the role of dysregulated STC in the progression of cancers of the reproductive system, including endometrial, cervical, and ovarian cancers. Additionally, the review evaluates the expression patterns and prognostic significance of STC in gynecological cancers by utilizing existing public datasets from The Cancer Genome Atlas to help decipher the multifaceted roles of these pleiotropic hormones in disease progression. Understanding the intricate mechanisms by which STC proteins influence all these reviewed conditions could lead to the development of targeted diagnostic and therapeutic strategies in the context of female reproductive health and oncology.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR—Cantonal Hospital of Fribourg and University of Fribourg, 79085 Fribourg, Switzerland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14152 Huddinge, Stockholm, Sweden
| |
Collapse
|
3
|
Panahipour L, Micucci C, Gelmetti B, Gruber R. In Vitro Bioassay for Damage-Associated Molecular Patterns Arising from Injured Oral Cells. Bioengineering (Basel) 2024; 11:687. [PMID: 39061769 PMCID: PMC11273541 DOI: 10.3390/bioengineering11070687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Gingival fibroblasts are a significant source of paracrine signals required to maintain periodontal homeostasis and to mediate pathological events linked to periodontitis and oral squamous cell carcinomas. Among the potential paracrine signals are stanniocalcin-1 (STC1), involved in oxidative stress and cellular survival; amphiregulin (AREG), a growth factor that mediates the cross-talk between immune cells and epithelial cells; chromosome 11 open reading frame 96 (C11orf96) with an unclear biologic function; and the inflammation-associated prostaglandin E synthase (PTGES). Gingival fibroblasts increasingly express these genes in response to bone allografts containing remnants of injured cells. Thus, the gene expression might be caused by the local release of damage-associated molecular patterns arising from injured cells. The aim of this study is consequently to use the established gene panel as a bioassay to measure the damage-associated activity of oral cell lysates. To this aim, we have exposed gingival fibroblasts to lysates prepared from the squamous carcinoma cell lines TR146 and HSC2, oral epithelial cells, and gingival fibroblasts. We report here that all lysates significantly increased the transcription of the entire gene panel, supported for STC1 at the protein level. Blocking TGF-β receptor 1 kinase with SB431542 only partially reduced the forced expression of STC1, AREG, and C11orf96. SB431542 even increased the PTGES expression. Together, these findings suggest that the damage signals originating from oral cells can change the paracrine activity of gingival fibroblasts. Moreover, the expression panel of genes can serve as a bioassay for testing the biocompatibility of materials for oral application.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.); (B.G.)
| | - Chiara Micucci
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.); (B.G.)
| | - Benedetta Gelmetti
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.); (B.G.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.); (B.G.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
4
|
Panahipour L, Abbasabadi AO, Wagner A, Kratochwill K, Pichler M, Gruber R. Bone Allograft Acid Lysates Change the Genetic Signature of Gingival Fibroblasts. Int J Mol Sci 2023; 24:16181. [PMID: 38003371 PMCID: PMC10671348 DOI: 10.3390/ijms242216181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Bone allografts are widely used as osteoconductive support to guide bone regrowth. Bone allografts are more than a scaffold for the immigrating cells as they maintain some bioactivity of the original bone matrix. Yet, it remains unclear how immigrating cells respond to bone allografts. To this end, we have evaluated the response of mesenchymal cells exposed to acid lysates of bone allografts (ALBA). RNAseq revealed that ALBA has a strong impact on the genetic signature of gingival fibroblasts, indicated by the increased expression of IL11, AREG, C11orf96, STC1, and GK-as confirmed by RT-PCR, and for IL11 and STC1 by immunoassays. Considering that transforming growth factor-β (TGF-β) is stored in the bone matrix and may have caused the expression changes, we performed a proteomics analysis, TGF-β immunoassay, and smad2/3 nuclear translocation. ALBA neither showed detectable TGF-β nor was the lysate able to induce smad2/3 translocation. Nevertheless, the TGF-β receptor type I kinase inhibitor SB431542 significantly decreased the expression of IL11, AREG, and C11orf96, suggesting that other agonists than TGF-β are responsible for the robust cell response. The findings suggest that IL11, AREG, and C11orf96 expression in mesenchymal cells can serve as a bioassay reflecting the bioactivity of the bone allografts.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
5
|
Wan HT, Ng AH, Lee WK, Shi F, Wong CKC. Identification and characterization of a membrane receptor that binds to human STC1. Life Sci Alliance 2022; 5:5/11/e202201497. [PMID: 35798563 PMCID: PMC9263378 DOI: 10.26508/lsa.202201497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
A study using TriCEPS-based ligand–receptor methodology and surface plasmon resonance assays identified that human stanniocalcin-1 binds to insulin-like growth factor-2 receptors in human leukemia monocytic cells with high affinity. Stanniocalcin-1 (STC1) is a hypocalcemic hormone originally identified in bony fishes. The mammalian homolog is found to be involved in inflammation and carcinogenesis, among other physiological functions. In this study, we used the TriCEPS-based ligand–receptor methodology to identify the putative binding proteins of human STC1 (hSTC1) in the human leukemia monocytic cell line, ThP-1. LC–MS/MS analysis of peptides from shortlisted hSTC1-binding proteins detected 32 peptides that belong to IGF2/MPRI. Surface plasmon resonance assay demonstrated that hSTC1 binds to immobilized IGF2R/MPRI with high affinity (10–20 nM) and capacity (Rmax 70–100%). The receptor binding data are comparable with those of (CREG) cellular repressor of E1A-stimulated gene a known ligand of IGF2R/MPRI, with Rmax of 75–80% and affinity values of 1–2 nM. The surface plasmon resonance competitive assays showed CREG competed with hSTC1 in binding to IGF2R/MPRI. The biological effects of hSTC1 on ThP-1 cells were demonstrated via IGF2R/MPRI to significantly reduce secreted levels of IL-1β. This is the first study to reveal the high-affinity binding of hSTC1 to the membrane receptor IGF2R/MPRI.
Collapse
Affiliation(s)
- Hin Ting Wan
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Alice Hm Ng
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wang Ka Lee
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Feng Shi
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chris Kong-Chu Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
6
|
Li S, Liu C, Goldstein A, Xin Y, Ke C, Duan C. Calcium State-Dependent Regulation of Epithelial Cell Quiescence by Stanniocalcin 1a. Front Cell Dev Biol 2021; 9:662915. [PMID: 33898465 PMCID: PMC8063699 DOI: 10.3389/fcell.2021.662915] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/08/2021] [Indexed: 11/15/2022] Open
Abstract
The molecular mechanisms regulating cell quiescence-proliferation balance are not well defined. Using a zebrafish model, we report that Stc1a, a secreted glycoprotein, plays a key role in regulating the quiescence-proliferation balance of Ca2+ transporting epithelial cells (ionocytes). Zebrafish stc1a, but not the other stc genes, is expressed in a Ca2+ state-dependent manner. Genetic deletion of stc1a, but not stc2b, increased ionocyte proliferation, leading to elevated body Ca2+ levels, cardiac edema, body swelling, and premature death. The increased ionocyte proliferation was accompanied by an increase in the IGF1 receptor-mediated PI3 kinase-Akt-Tor signaling activity in ionocytes. Inhibition of the IGF1 receptor, PI3 kinase, Akt, and Tor signaling reduced ionocyte proliferation and rescued the edema and premature death in stc1a–/– fish, suggesting that Stc1a promotes ionocyte quiescence by suppressing local IGF signaling activity. Mechanistically, Stc1 acts by inhibiting Papp-aa, a zinc metalloproteinase degrading Igfbp5a. Inhibition of Papp-aa proteinase activity restored ionocyte quiescence-proliferation balance. Genetic deletion of papp-aa or its substrate igfbp5a in the stc1a–/– background reduced ionocyte proliferation and rescued the edema and premature death. These findings uncover a novel and Ca2+ state-dependent pathway regulating cell quiescence. Our findings also provide new insights into the importance of ionocyte quiescent-proliferation balance in organismal Ca2+ homeostasis and survival.
Collapse
Affiliation(s)
- Shuang Li
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Chengdong Liu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Allison Goldstein
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Yi Xin
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Caihuan Ke
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
7
|
Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, You L, Zheng L, Zhang T, Zhao Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J Cell Mol Med 2020; 24:7686-7696. [PMID: 32468698 PMCID: PMC7348177 DOI: 10.1111/jcmm.15348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein stanniocalcin-1 functions as a regulatory endocrine hormone that maintains the balance of calcium and phosphorus in bony fish and as a paracrine/autocrine factor involved in many physiological/pathological processes in humans, including carcinogenesis. In this review, we provide an overview of (a) the possible mechanisms through which STC1 affects the malignant properties of cancer, (b) transcriptional and post-transcriptional regulation pathways of STC1 and (c) the potential clinical relevance of STC1 as a cancer biomarker and even a therapeutic target in the future. Exploring the role of STC1 in cancer development may provide a better understanding of the tumorigenesis process in humans and may facilitate finding an effective therapeutic method against cancer.
Collapse
Affiliation(s)
- Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Raimondo D, Remoli C, Astrologo L, Burla R, La Torre M, Vernì F, Tagliafico E, Corsi A, Del Giudice S, Persichetti A, Giannicola G, Robey PG, Riminucci M, Saggio I. Changes in gene expression in human skeletal stem cells transduced with constitutively active Gsα correlates with hallmark histopathological changes seen in fibrous dysplastic bone. PLoS One 2020; 15:e0227279. [PMID: 31999703 PMCID: PMC6991960 DOI: 10.1371/journal.pone.0227279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/16/2019] [Indexed: 02/05/2023] Open
Abstract
Fibrous dysplasia (FD) of bone is a complex disease of the skeleton caused by dominant activating mutations of the GNAS locus encoding for the α subunit of the G protein-coupled receptor complex (Gsα). The mutation involves a substitution of arginine at position 201 by histidine or cysteine (GsαR201H or R201C), which leads to overproduction of cAMP. Several signaling pathways are implicated downstream of excess cAMP in the manifestation of disease. However, the pathogenesis of FD remains largely unknown. The overall FD phenotype can be attributed to alterations of skeletal stem/progenitor cells which normally develop into osteogenic or adipogenic cells (in cis), and are also known to provide support to angiogenesis, hematopoiesis, and osteoclastogenesis (in trans). In order to dissect the molecular pathways rooted in skeletal stem/progenitor cells by FD mutations, we engineered human skeletal stem/progenitor cells with the GsαR201C mutation and performed transcriptomic analysis. Our data suggest that this FD mutation profoundly alters the properties of skeletal stem/progenitor cells by pushing them towards formation of disorganized bone with a concomitant alteration of adipogenic differentiation. In addition, the mutation creates an altered in trans environment that induces neovascularization, cytokine/chemokine changes and osteoclastogenesis. In silico comparison of our data with the signature of FD craniofacial samples highlighted common traits, such as the upregulation of ADAM (A Disintegrin and Metalloprotease) proteins and other matrix-related factors, and of PDE7B (Phosphodiesterase 7B), which can be considered as a buffering process, activated to compensate for excess cAMP. We also observed high levels of CEBPs (CCAAT-Enhancer Binding Proteins) in both data sets, factors related to browning of white fat. This is the first analysis of the reaction of human skeletal stem/progenitor cells to the introduction of the FD mutation and we believe it provides a useful background for further studies on the molecular basis of the disease and for the identification of novel potential therapeutic targets.
Collapse
Affiliation(s)
- Domenico Raimondo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cristina Remoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Letizia Astrologo
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Mattia La Torre
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Enrico Tagliafico
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Simona Del Giudice
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Agnese Persichetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Giannicola
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedics Sciences, Sapienza University of Rome, Rome, Italy
| | - Pamela G. Robey
- National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, United States of America
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- * E-mail: (IS); (MR)
| | - Isabella Saggio
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
- School of Biological Sciences, NTU Institute of Structural Biology, Nanyang Technological University, Singapore
- * E-mail: (IS); (MR)
| |
Collapse
|
9
|
Asano N, Takeshima H, Yamashita S, Takamatsu H, Hattori N, Kubo T, Yoshida A, Kobayashi E, Nakayama R, Matsumoto M, Nakamura M, Ichikawa H, Kawai A, Kondo T, Ushijima T. Epigenetic reprogramming underlies efficacy of DNA demethylation therapy in osteosarcomas. Sci Rep 2019; 9:20360. [PMID: 31889115 PMCID: PMC6937291 DOI: 10.1038/s41598-019-56883-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma (OS) patients with metastasis or recurrent tumors still suffer from poor prognosis. Studies have indicated the efficacy of DNA demethylation therapy for OS, but the underlying mechanism is still unclear. Here, we aimed to clarify the mechanism of how epigenetic therapy has therapeutic efficacy in OS. Treatment of four OS cell lines with a DNA demethylating agent, 5-aza-2′-deoxycytidine (5-aza-dC) treatment, markedly suppressed their growth, and in vivo efficacy was further confirmed using two OS xenografts. Genome-wide DNA methylation analysis showed that 10 of 28 primary OS had large numbers of methylated CpG islands while the remaining 18 OS did not, clustering together with normal tissue samples and Ewing sarcoma samples. Among the genes aberrantly methylated in primary OS, genes involved in skeletal system morphogenesis were present. Searching for methylation-silenced genes by expression microarray screening of two OS cell lines after 5-aza-dC treatment revealed that multiple tumor-suppressor and osteo/chondrogenesis-related genes were re-activated by 5-aza-dC treatment of OS cells. Simultaneous activation of multiple genes related to osteogenesis and cell proliferation, namely epigenetic reprogramming, was considered to underlie the efficacy of DNA demethylation therapy in OS.
Collapse
Affiliation(s)
- Naofumi Asano
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Takeshima
- Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Satoshi Yamashita
- Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hironori Takamatsu
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takashi Kubo
- Department of Clinical Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akihiko Yoshida
- Department of Pathology and Clinical Laboratory, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Eisuke Kobayashi
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Robert Nakayama
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hitoshi Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
10
|
de las Heras-Saldana S, Clark SA, Duijvesteijn N, Gondro C, van der Werf JHJ, Chen Y. Combining information from genome-wide association and multi-tissue gene expression studies to elucidate factors underlying genetic variation for residual feed intake in Australian Angus cattle. BMC Genomics 2019; 20:939. [PMID: 31810463 PMCID: PMC6898931 DOI: 10.1186/s12864-019-6270-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/07/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) are extensively used to identify single nucleotide polymorphisms (SNP) underlying the genetic variation of complex traits. However, much uncertainly often still exists about the causal variants and genes at quantitative trait loci (QTL). The aim of this study was to identify QTL associated with residual feed intake (RFI) and genes in these regions whose expression is also associated with this trait. Angus cattle (2190 steers) with RFI records were genotyped and imputed to high density arrays (770 K) and used for a GWAS approach to identify QTL associated with RFI. RNA sequences from 126 Angus divergently selected for RFI were analyzed to identify the genes whose expression was significantly associated this trait with special attention to those genes residing in the QTL regions. RESULTS The heritability for RFI estimated for this Angus population was 0.3. In a GWAS, we identified 78 SNPs associated with RFI on six QTL (on BTA1, BTA6, BTA14, BTA17, BTA20 and BTA26). The most significant SNP was found on chromosome BTA20 (rs42662073) and explained 4% of the genetic variance. The minor allele frequencies of significant SNPs ranged from 0.05 to 0.49. All regions, except on BTA17, showed a significant dominance effect. In 1 Mb windows surrounding the six significant QTL, we found 149 genes from which OAS2, STC2, SHOX, XKR4, and SGMS1 were the closest to the most significant QTL on BTA17, BTA20, BTA1, BTA14, and BTA26, respectively. In a 2 Mb windows around the six significant QTL, we identified 15 genes whose expression was significantly associated with RFI: BTA20) NEURL1B and CPEB4; BTA17) RITA1, CCDC42B, OAS2, RPL6, and ERP29; BTA26) A1CF, SGMS1, PAPSS2, and PTEN; BTA1) MFSD1 and RARRES1; BTA14) ATP6V1H and MRPL15. CONCLUSIONS Our results showed six QTL regions associated with RFI in a beef Angus population where five of these QTL contained genes that have expression associated with this trait. Therefore, here we show that integrating information from gene expression and GWAS studies can help to better understand the genetic mechanisms that determine variation in complex traits.
Collapse
Affiliation(s)
| | - Samuel A. Clark
- School of Environmental and Rural Science, University of New England, Armidale, NSW Australia
| | - Naomi Duijvesteijn
- School of Environmental and Rural Science, University of New England, Armidale, NSW Australia
| | - Cedric Gondro
- School of Environmental and Rural Science, University of New England, Armidale, NSW Australia
- Department of Animal Science, Michigan State University, East Lansing, MI USA
| | | | - Yizhou Chen
- Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, NSW Australia
| |
Collapse
|
11
|
Nawrocki MJ, Perek B, Sujka-Kordowska P, Konwerska A, Kałużna S, Zawierucha P, Bruska M, Zabel M, Jemielity M, Nowicki M, Kempisty B, Malińska A. Differences in Expression of Genes Involved in Bone Development and Morphogenesis in the Walls of Internal Thoracic Artery and Saphenous Vein Conduits May Provide Markers Useful for Evaluation Graft Patency. Int J Mol Sci 2019; 20:ijms20194890. [PMID: 31581653 PMCID: PMC6801533 DOI: 10.3390/ijms20194890] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 01/22/2023] Open
Abstract
Coronary artery bypass grafting (CABG) is one of the most efficient procedures for patients with advanced coronary artery disease. From all the blood vessels with the potential to be used in this procedure, the internal thoracic artery (ITA) and the saphenous vein (SV) are the most commonly applied as aortocoronary conduits. Nevertheless, in order to evaluate the graft patency and efficiency effectively, basic knowledge should be constantly expanding at the molecular level as well, as the understanding of predictive factors is still limited. In this study, we have employed the expressive microarray approach, validated with Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR), to analyze the transcriptome of both venous and arterial grafts. Searching for potential molecular factors, we analyzed differentially expressed gene ontologies involved in bone development and morphogenesis, for the possibility of discovery of new markers for the evaluation of ITA and SV segment quality. Among three ontological groups of interest—“endochondral bone morphogenesis”, “ossification”, and “skeletal system development”—we found six genes common to all of them. BMP6, SHOX2, COL13A1, CSGALNACT1, RUNX2, and STC1 showed differential expression patterns in both analyzed vessels. STC1 and COL13A1 were upregulated in ITA samples, whereas others were upregulated in SV. With regard to the Runx2 protein function in osteogenic phenotype regulation, the RUNX2 gene seems to be of paramount importance in assessing the potential of ITA, SV, and other vessels used in the CABG procedure. Overall, the presented study provided valuable insight into the molecular background of conduit characterization, and thus indicated genes that may be the target of subsequent studies, also at the protein level. Moreover, it has been suggested that RUNX2 may be recognized as a molecular marker of osteogenic changes in human blood vessels.
Collapse
Affiliation(s)
- Mariusz J Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland.
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Aneta Konwerska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Sandra Kałużna
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Piotr Zawierucha
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Małgorzata Bruska
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland.
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wrocław, Poland.
| | - Marek Jemielity
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic.
| | - Agnieszka Malińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland.
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland.
| |
Collapse
|
12
|
Sundelacruz S, Moody AT, Levin M, Kaplan DL. Membrane Potential Depolarization Alters Calcium Flux and Phosphate Signaling During Osteogenic Differentiation of Human Mesenchymal Stem Cells. Bioelectricity 2019; 1:56-66. [PMID: 32292891 PMCID: PMC6524654 DOI: 10.1089/bioe.2018.0005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Membrane potential (Vmem) changes accompany important events in embryonic development and organ regeneration. Recent studies have pointed to its function as a potent regulator of cell proliferation, differentiation, migration, and tissue regeneration. We have previously reported that Vmem depolarization and hyperpolarization control the osteogenic (OS) differentiation potential of human mesenchymal stem cells (hMSCs). Materials and Methods: In this study, we sought to understand the mechanism(s) underlying voltage regulation of hMSC differentiation. We investigated the role of calcium and phosphate ion flux in the depolarization response of OS-differentiating hMSCs, as these ions are the two major inorganic components of the bone mineral matrix and are indicative of mature osteoblast function. Results: Our results suggest that inorganic phosphate levels play a larger role than calcium flux in mediating hMSC response to depolarization and that the expression of stanniocalcin 1 (STC1), a protein that regulates calcium and phosphate homeostasis in osteoblasts, is functionally required for the depolarization response during the early stages of differentiation. Conclusion: Depolarization alters hMSC differentiation through a phosphate signaling pathway involving STC1. This study enriches our mechanistic understanding of hMSC response to endogenous voltage cues.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Amy Thurber Moody
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Medford, Massachusetts
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
13
|
Fujimoto M, Hwa V, Dauber A. Novel Modulators of the Growth Hormone - Insulin-Like Growth Factor Axis: Pregnancy-Associated Plasma Protein-A2 and Stanniocalcin-2. J Clin Res Pediatr Endocrinol 2017; 9:1-8. [PMID: 29280739 PMCID: PMC5790331 DOI: 10.4274/jcrpe.2017.s001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Growth hormone (GH) and its mediator, insulin-like growth factor-1 (IGF-1), play a critical role in human growth. In circulation, IGF-1 is found in a ternary complex with IGF binding proteins (IGFBPs) and acid labile subunit (ALS) but little attention has been paid to the regulation of IGF-1 bioavailability. Recently, pregnancy-associated plasma protein-A2 (PAPP-A2) and stanniocalcin-2 (STC2) were identified as novel modulators of IGF-I bioavailability. PAPP-A2 is a protease which cleaves IGFBP-3 and -5, while STC2 inhibits PAPP-A and PAPP-A2 activity. In collaboration with a group in Madrid, we reported the first human cases carrying mutations in the PAPPA2 gene who presented with short stature, elevated total IGF-1, IGFBP-3, IGFBP-5 and ALS, but low free IGF-1. Additionally, the patients demonstrated insulin resistance and below average bone mineral density (BMD). The PAPP-A2 deficient patients were treated with recombinant human IGF-1, resulting in improvements in growth velocity, insulin resistance, and BMD. These findings suggested that the bioactive, free IGF-1 liberated from IGFBPs by PAPP-A2 is important for human growth. Mouse models of PAPP-A2 and STC2 provide further insights into their roles in growth physiology. This review will summarize new insights into PAPP-A2 and STC2 and their role in the GH-IGF axis, thereby highlighting the importance of the regulation of IGF-1 bioavailability in human health and disease.
Collapse
Affiliation(s)
- Masanobu Fujimoto
- Cincinnati Children’s Hospital Medical Center, Cincinnati Center for Growth Disorders, Clinic of Endocrinology, Cincinnati, Ohio, USA
| | - Vivian Hwa
- Cincinnati Children’s Hospital Medical Center, Cincinnati Center for Growth Disorders, Clinic of Endocrinology, Cincinnati, Ohio, USA
| | - Andrew Dauber
- Cincinnati Children’s Hospital Medical Center, Cincinnati Center for Growth Disorders, Clinic of Endocrinology, Cincinnati, Ohio, USA
| |
Collapse
|
14
|
Leung CC, Wong CK. Effects of STC1 overexpression on tumorigenicity and metabolism of hepatocellular carcinoma. Oncotarget 2017; 9:6852-6861. [PMID: 29467934 PMCID: PMC5805520 DOI: 10.18632/oncotarget.23566] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/28/2017] [Indexed: 12/25/2022] Open
Abstract
Stanniocalcin-1 (STC1) is a paracrine factor associated with inflammation and carcinogenesis. Using clinicopathological data, we previously reported that a greater expression of STC1 in hepatocellular carcinoma (HCC) was significantly correlated with smaller tumor size. The underlying mechanism on the correlation is not known. In this study, using a metastatic HCC cell-line (MHCC-97L, P) and lentiviral vector mediated-STC1 overexpression, the inoculation of STC1-overexpressing MHCC-97L (S1) cells in a nude mice xenograft model demonstrated reductions in tumor mass and volume. As compared with P cells, S1 cells exhibited epithelial phenotype with significantly lower plating efficiency and reduced migratory and proliferative potential. Using coulter counter for cell-sizing, S1 cells (17.6 μm) were significantly smaller than P cells (19.6 μm). Western blot analysis revealed that S1 cells exhibited reduced expression level of phosphorylated ribosomal protein S6 (p-rpS6). Moreover, an inhibition of the upstream kinase p70S6K was evident with the dephosphorylation of Thr389 in the linker domain of the kinase. The inhibition of p70S6K/p-rpS6 pathway was accompanied with reduced cellular ATP level and increase of p-AMPK in S1 cells. Significantly lower rates of glycolysis and extracellular O2 consumption in S1 cells exhibited a lower cellular energy status. Since a faster rate of ATP production is essential to support cancer growth and metastasis, the present study identified the effect of STC1-overexpression on reducing energy metabolism, leading to an activation of AMPK pathway but an inhibition of p70S6K/p-rpS6 signaling to reduce tumor growth.
Collapse
Affiliation(s)
- Cherry Ct Leung
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chris Kc Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
15
|
Chen JL, Zou C, Chen Y, Zhu W, Liu W, Huang J, Liu Q, Wang D, Duan L, Xiong J, Cui J, Jia Z, Wang D. TGFβ1 induces hypertrophic change and expression of angiogenic factors in human chondrocytes. Oncotarget 2017; 8:91316-91327. [PMID: 29207646 PMCID: PMC5710926 DOI: 10.18632/oncotarget.20509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/04/2017] [Indexed: 11/25/2022] Open
Abstract
The transforming growth factor β1 (TGFβ1) plays an important role in cartilage development. However, whether TGFβ1 stimulates chondrocyte proliferation and cartilage regeneration in osteoarthritis (OA) remains elusive, especially in the context of different treatment and tissue environments. In the present study, we investigated the role of TGFβ1 in human chondrocyte culture in vitro, focusing on the morphological change of chondrocytes and the expression of angiogenic factors upon TGFβ1 stimulation. We found increased expression of biomarkers indicating chondrocyte hypertrophy and the chondrocytes aggregated to form networks when they were treated with TGFβ1. DNA microarray analysis revealed significantly increased expression of genes related to blood vessel formation in TGFβ1 treatment group compared to control group. Matrigel assay further demonstrated that chondrocytes had the potential to form network-like structure. These results suggested that TGFβ1 induces the hypertrophic change of chondrocytes culture in vitro and induce expression of angiogenic biomarkers. Therefore, application of TGFβ1 for chondrocyte culture in practice should be considered prudentially and targeting TGFβ1 or relevant receptors to block the signaling pathway might be a strategy to prevent or alleviate progression of osteoarthritis.
Collapse
Affiliation(s)
- Jie-Lin Chen
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Chang Zou
- Shenzhen Public Service Platform for Cancer Precision Medicine and Molecular Diagnosis, Shenzhen 518020, China.,Clinical Medical Research Center, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, 518020 China
| | - Yunfang Chen
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518035, Guangdong Province, China
| | - Weimin Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Wei Liu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Jianghong Huang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Qisong Liu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Daming Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Jianyi Xiong
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Jiaming Cui
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Zhaofeng Jia
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China.,Shenzhen Centre for Sports Medicine and Engineering Technology, Shenzhen 518035, Guangdong Province, China
| |
Collapse
|
16
|
Lin CH, Hu HJ, Hwang PP. Molecular Physiology of the Hypocalcemic Action of Fibroblast Growth Factor 23 in Zebrafish (Danio rerio). Endocrinology 2017; 158:1347-1358. [PMID: 28323996 DOI: 10.1210/en.2016-1883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/23/2017] [Indexed: 12/23/2022]
Abstract
Fibroblast growth factor 23 (FGF23), a hormone required for phosphorus metabolism, was recently proposed to act on Ca2+ uptake; however, the available evidence of how FGF23 controls the body fluid Ca2+ homeostasis needs to be further clarified. The use of zebrafish as a model system revealed that FGF23 is specifically expressed in the corpuscles of Stannius (CS), an organ involved in Ca2+ homeostasis in fish, and that its expression is stimulated by ambient water with a high Ca2+ level. The overexpression of FGF23 inhibited Ca2+ uptake by downregulating the messenger RNA (mRNA) expression of epithelium calcium channel. Calcium-sensing receptor (CaSR), which senses changes in extracellular Ca2+ levels and modulates calciotropic hormones in organs controlling Ca2+ homeostasis in vertebrates, was found to be coexpressed with FGF23 in the CS. In addition, upregulated expression of FGF23 mRNA was detected in morphants of stanniocalcin 1 (stc1, another hypocalcemic factor synthesized in the CS), and knockdown of CaSR suppressed such upregulation and enhanced Ca2+ uptake. Taken together, our data indicate that FGF23 functions as a hypocalcemic hormone in zebrafish and that the CaSR/STC1-FGF23 axis is involved in body fluid Ca2+ homeostasis in vertebrates.
Collapse
Affiliation(s)
- Chia-Hao Lin
- National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Huei-Jyun Hu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan, ROC
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan, ROC
| |
Collapse
|
17
|
Juhanson P, Rull K, Kikas T, Laivuori H, Vaas P, Kajantie E, Heinonen S, Laan M. Stanniocalcin-1 Hormone in Nonpreeclamptic and Preeclamptic Pregnancy: Clinical, Life-Style, and Genetic Modulators. J Clin Endocrinol Metab 2016; 101:4799-4807. [PMID: 27603899 PMCID: PMC5155696 DOI: 10.1210/jc.2016-1873] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/31/2016] [Indexed: 11/19/2022]
Abstract
CONTEXT AND OBJECTIVES The study represents the first comprehensive analysis of Stanniocalcin-1 (STC1) hormone in human pregnancy, assessing clinical, lifestyle, and genetic determinants of circulating STC1 at term. DESIGN, SETTING, AND PARTICIPANTS Participants included women with (n = 50) and without (n = 316) preeclampsia (PE) at delivery, recruited in the REPROgrammed fetal and/or maternal METAbolism (REPROMETA) study (2006-2011, Estonia). Genetic association analysis combined PE cases (n = 597) and controls (n = 623) from the REPROMETA and Finnish Genetics of Preeclampsia Consortium (2008-2011) studies. MAIN OUTCOME MEASURE(S) Maternal postpartum plasma STC1 was measured by ELISA (n = 366) and placental STC1 gene expression by TaqMan quantitative RT-PCR (n = 120). Genotyping was performed using Sequenom MassArray. RESULTS Significantly higher STC1 plasma level was measured for the PE (median, 1952 pg/mL; 1030-4284 pg/mL) compared with non-PE group (median, 1562 pg/mL; 423-3781 pg/mL; P = 3.7 × 10-4, Mann-Whitney U test). Statistical significance was enhanced after adjustment for cofactors (linear regression, P = 1.8 × 10-6). STC1 measurements were negatively correlated with maternal smoking. Prepregnancy body mass index had a positive correlation with STC1 only among PE patients (r = 0.45; P = .001). The strongest genetic association with hormone concentrations was detected for STC1 single nucleotide polymorphisms rs3758089 (C allele: minor allele frequency, 5%; linear regression: β = 249.2 pg/mL; P = .014) and rs12678447 (G allele: minor allele frequency, 7%; β = 147.0 pg/mL; P = .082). rs12678447 placental genotypes were significantly associated with STC1 gene expression (P = .014). The REPROMETA/Finnish Genetics of Preeclampsia Consortium meta-analysis suggested an increased risk to develop late-onset PE for the rs12678447 G allele carriers (P = .05; odds ratio = 1.38 [0.98-1.93]). CONCLUSIONS Increased STC1 hormone represents a hallmark of late-onset PE. STC1 gene variants modulate placental gene expression and maternal hormone levels.
Collapse
Affiliation(s)
- Peeter Juhanson
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Kristiina Rull
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Triin Kikas
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Hannele Laivuori
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Pille Vaas
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Eero Kajantie
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Seppo Heinonen
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| | - Maris Laan
- Human Molecular Genetics Research Group (P.J., K.R., T.K., M.L.), Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia; Department of Obstetrics and Gynaecology (K.R., P.V.), University of Tartu, and Women's Clinic of Tartu University Hospital (K.R., P.V.), Tartu 51014, Estonia; Medical and Clinical Genetics (H.L.), University of Helsinki and Helsinki University Hospital, and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, FIN-00014 Helsinki, Finland; Obstetrics and Gynecology (H.L., S.H.) and Children's Hospital (E.K.), Helsinki University Hospital and University of Helsinki, FIN-00029 Helsinki, Finland; Chronic Disease Prevention Unit (E.K.), National Institute for Health and Welfare, FIN-00271 Helsinki, Finland; Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (E.K.), Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FIN-90014 Oulu, Finland; and Institute of Biomedicine and Translational Medicine (M.L.), University of Tartu, Tartu 50411, Estonia
| |
Collapse
|
18
|
Dotterweich J, Schlegelmilch K, Keller A, Geyer B, Schneider D, Zeck S, Tower RJJ, Ebert R, Jakob F, Schütze N. Contact of myeloma cells induces a characteristic transcriptome signature in skeletal precursor cells -Implications for myeloma bone disease. Bone 2016; 93:155-166. [PMID: 27519972 DOI: 10.1016/j.bone.2016.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/24/2016] [Accepted: 08/08/2016] [Indexed: 10/21/2022]
Abstract
Physical interaction of skeletal precursors with multiple myeloma cells has been shown to suppress their osteogenic potential while favoring their tumor-promoting features. Although several transcriptome analyses of myeloma patient-derived mesenchymal stem cells have displayed differences compared to their healthy counterparts, these analyses insufficiently reflect the signatures mediated by tumor cell contact, vary due to different methodologies, and lack results in lineage-committed precursors. To determine tumor cell contact-mediated changes on skeletal precursors, we performed transcriptome analyses of mesenchymal stem cells and osteogenic precursor cells cultured in contact with the myeloma cell line INA-6. Comparative analyses confirmed dysregulation of genes which code for known disease-relevant factors and additionally revealed upregulation of genes that are associated with plasma cell homing, adhesion, osteoclastogenesis, and angiogenesis. Osteoclast-derived coupling factors, a dysregulated adipogenic potential, and an imbalance in favor of anti-anabolic factors may play a role in the hampered osteoblast differentiation potential of mesenchymal stem cells. Angiopoietin-Like 4 (ANGPTL4) was selected from a list of differentially expressed genes as a myeloma cell contact-dependent target in skeletal precursor cells which warranted further functional analyses. Adhesion assays with full-length ANGPTL4-coated plates revealed a potential role of this protein in INA-6 cell attachment. This study expands knowledge of the myeloma cell contact-induced signature in the stromal compartment of myelomatous bones and thus offers potential targets that may allow detection and treatment of myeloma bone disease at an early stage.
Collapse
Affiliation(s)
- Julia Dotterweich
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Katrin Schlegelmilch
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Alexander Keller
- DNA-Analytics Core Facility, Biocenter and Department of Animal Ecology and Tropical Biology, University of Würzburg, Würzburg, Germany
| | - Beate Geyer
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Doris Schneider
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Robert J J Tower
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany.
| | - Norbert Schütze
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Ohkouchi S, Ono M, Kobayashi M, Hirano T, Tojo Y, Hisata S, Ichinose M, Irokawa T, Ogawa H, Kurosawa H. Myriad Functions of Stanniocalcin-1 (STC1) Cover Multiple Therapeutic Targets in the Complicated Pathogenesis of Idiopathic Pulmonary Fibrosis (IPF). CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2015; 9:91-6. [PMID: 26740747 PMCID: PMC4696838 DOI: 10.4137/ccrpm.s23285] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/14/2015] [Accepted: 11/05/2015] [Indexed: 12/29/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an intractable disease for which the pathological findings are characterized by temporal and spatial heterogeneity. The pathogenesis is composed of myriad factors, including repetitive injuries to epithelial cells, alterations in immunity, the formation of vascular leakage and coagulation, abnormal wound healing, fibrogenesis, and collagen accumulation. Therefore, the molecular target drugs that are used or attempted for treatment or clinical trials may not cover the myriad therapeutic targets of IPF. In addition, the complicated pathogenesis results in a lack of informative biomarkers to diagnose accurately the status of IPF. These facts point out the necessity of using a combination of drugs, that is, each single drug with molecular targets or a single drug with multiple therapeutic targets. In this review, we introduce a humoral factor, stanniocalcin-1 (STC1), which has myriad functions, including the maintenance of calcium homeostasis, the promotion of early wound healing, uncoupling respiration (aerobic glycolysis), reepithelialization in damaged tissues, the inhibition of vascular leakage, and the regulation of macrophage functions to keep epithelial and endothelial homeostasis, which may adequately cover the myriad therapeutic targets of IPF.
Collapse
Affiliation(s)
- Shinya Ohkouchi
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Occupational Health, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Manabu Ono
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Makoto Kobayashi
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Taizou Hirano
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yutaka Tojo
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Shu Hisata
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan; Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Toshiya Irokawa
- Department of Occupational Health, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hiromasa Ogawa
- Department of Occupational Health, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hajime Kurosawa
- Department of Occupational Health, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
20
|
Cellular processes involved in human epidermal cells exposed to extremely low frequency electric fields. Cell Signal 2015; 27:889-98. [DOI: 10.1016/j.cellsig.2015.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/08/2015] [Indexed: 01/18/2023]
|
21
|
Terra SR, Cardoso JCR, Félix RC, Martins LAM, Souza DOG, Guma FCR, Canário AVM, Schein V. STC1 interference on calcitonin family of receptors signaling during osteoblastogenesis via adenylate cyclase inhibition. Mol Cell Endocrinol 2015; 403:78-87. [PMID: 25591908 DOI: 10.1016/j.mce.2015.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/15/2014] [Accepted: 01/06/2015] [Indexed: 12/24/2022]
Abstract
Stanniocalcin 1 (STC1) and calcitonin gene-related peptide (CGRP) are involved in bone formation/remodeling. Here we investigate the effects of STC1 on functional heterodimer complex CALCRL/RAMP1, expression and activity during osteoblastogenesis. STC1 did not modify CALCRL and ramp1 gene expression during osteoblastogenesis when compared to controls. However, plasma membrane spatial distribution of CALCRL/RAMP1 was modified in 7-day pre-osteoblasts exposed to either CGRP or STC1, and both peptides induced CALCRL and RAMP1 assembly. CGRP, but not STC1 stimulated cAMP accumulation in 7-day osteoblasts and in CALCRL/RAMP1 transfected HEK293 cells. Furthermore, STC1 inhibited forskolin stimulated cAMP accumulation of HEK293 cells, but not in CALCRL/RAMP1 transfected HEK293 cells. However, STC1 inhibited cAMP accumulation in calcitonin receptor (CTR) HEK293 transfected cells stimulated by calcitonin. In conclusion, STC1 signals through inhibitory G-protein modulates CGRP receptor spatial localization during osteoblastogenesis and may function as a regulatory factor interacting with calcitonin peptide members during bone formation.
Collapse
Affiliation(s)
- Silvia R Terra
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90003-035, Brazil
| | - João Carlos R Cardoso
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro 8005-139, Portugal
| | - Rute C Félix
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro 8005-139, Portugal
| | - Leo Anderson M Martins
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90003-035, Brazil
| | - Diogo Onofre G Souza
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90003-035, Brazil
| | - Fatima C R Guma
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90003-035, Brazil
| | - Adelino Vicente M Canário
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro 8005-139, Portugal
| | - Vanessa Schein
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90003-035, Brazil; Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro 8005-139, Portugal.
| |
Collapse
|
22
|
Chou MY, Lin CH, Chao PL, Hung JC, Cruz SA, Hwang PP. Stanniocalcin-1 controls ion regulation functions of ion-transporting epithelium other than calcium balance. Int J Biol Sci 2015; 11:122-32. [PMID: 25561895 PMCID: PMC4279088 DOI: 10.7150/ijbs.10773] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/18/2014] [Indexed: 01/08/2023] Open
Abstract
Stanniocalcin-1 (STC-1) was first identified to involve in Ca(2+) homeostasis in teleosts, and was thought to act as a hypocalcemic hormone in vertebrate. Recent studies suggested that STC-1 exhibits broad effects on ion balance, not confines to Ca(2+), but the mechanism of this regulation process remains largely unknown. Here, we used zebrafish embryos as an alternative in vivo model to investigate how STC-1 regulates transepithelial ion transport function in ion-transporting epithelium. Expression of stc-1 mRNA in zebrafish embryos was increased in high-Ca(2+) environments but decreased by acidic and ion-deficient treatments while overexpression of stc-1 impaired the hypotonic acclimation by decreasing whole body Ca(2+), Na(+), and Cl(-) contents and H(+) secretion ability. Injection of STC-1 mRNA also down-regulated mRNA expressions of epithelial Ca(2+) channel, H(+)-ATPase, and Na(+)-Cl(-) cotransporter, suggesting the roles of STC-1 in regulation of ions other than Ca(2+). Knockdown of STC-1 caused an increase in ionocyte progenitors (foxi3a as the marker) and mature ionocytes (ion transporters as the markers), but did not affect epithelium stem cells (p63 as the marker) in the embryonic skin. Overexpression of STC-1 had the corresponding opposite effect on ionocyte progenitors, mature ionocytes in the embryonic skin. Taken together, STC-1 negatively regulates the number of ionocytes to reduce ionocyte functions. This process is important for body fluid ionic homeostasis, which is achieved by the regulation of ion transport functions in ionocytes. The present findings provide new insights into the broader functions of STC-1, a hypocalcemic hormone.
Collapse
Affiliation(s)
- Ming-Yi Chou
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan. ; 2. RIKEN Brain Science Institute, Laboratory for Developmental Gene Regulation, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Chia-Hao Lin
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Pei-Lin Chao
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Jo-Chi Hung
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Shelly A Cruz
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Pung-Pung Hwang
- 1. Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
23
|
Rondeau G, Abedinpour P, Desai P, Baron VT, Borgstrom P, Welsh J. Effects of different tissue microenvironments on gene expression in breast cancer cells. PLoS One 2014; 9:e101160. [PMID: 25004123 PMCID: PMC4086928 DOI: 10.1371/journal.pone.0101160] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/02/2014] [Indexed: 01/01/2023] Open
Abstract
In metastasis, circulating tumor cells penetrate the walls of blood vessels and enter the metastatic target tissue, thereby becoming exposed to novel and relatively unsupportive microenvironments. In the new microenvironments, the tumor cells often remain in a dormant state indefinitely and must adapt before they are able to successfully colonize the tissue. Very little is known about this adaptive process. We studied temporal changes in gene expression when breast cancer cells adapt to survive and grow on brain, bone marrow, and lung tissue maintained in an in vivo culture system, as models of the metastatic colonization of these tissues. We observed the transient activation of genes typically associated with homeostasis and stress during the initial stages of adaptation, followed by the activation of genes that mediate more advanced functions, such as elaboration of cell morphology and cell division, as the cells adapted to thrive in the host tissue microenvironment. We also observed the temporary induction of genes characteristic of the host tissue, which was particularly evident when tumor cells were grown on brain tissue. These early transient gene expression events suggest potential points of therapeutic intervention that are not evident in data from well-established tumors.
Collapse
Affiliation(s)
- Gaelle Rondeau
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - Parisa Abedinpour
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - Prerak Desai
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - Veronique T. Baron
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - Per Borgstrom
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
| | - John Welsh
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
24
|
Ramayo-Caldas Y, Fortes MRS, Hudson NJ, Porto-Neto LR, Bolormaa S, Barendse W, Kelly M, Moore SS, Goddard ME, Lehnert SA, Reverter A. A marker-derived gene network reveals the regulatory role of PPARGC1A, HNF4G, and FOXP3 in intramuscular fat deposition of beef cattle. J Anim Sci 2014; 92:2832-45. [PMID: 24778332 DOI: 10.2527/jas.2013-7484] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
High intramuscular fat (IMF) awards price premiums to beef producers and is associated with meat quality and flavor. Studying gene interactions and pathways that affect IMF might unveil causative physiological mechanisms and inform genomic selection, leading to increased accuracy of predictions of breeding value. To study gene interactions and pathways, a gene network was derived from genetic markers associated with direct measures of IMF, other fat phenotypes, feedlot performance, and a number of meat quality traits relating to body conformation, development, and metabolism that might be plausibly expected to interact with IMF biology. Marker associations were inferred from genomewide association studies (GWAS) based on high density genotypes and 29 traits measured on 10,181 beef cattle animals from 3 breed types. For the network inference, SNP pairs were assessed according to the strength of the correlation between their additive association effects across the 29 traits. The co-association inferred network was formed by 2,434 genes connected by 28,283 edges. Topological network parameters suggested a highly cohesive network, in which the genes are strongly functionally interconnected. Pathway and network analyses pointed towards a trio of transcription factors (TF) as key regulators of carcass IMF: PPARGC1A, HNF4G, and FOXP3. Importantly, none of these genes would have been deemed as significantly associated with IMF from the GWAS. Instead, a total of 313 network genes show significant co-association with the 3 TF. These genes belong to a wide variety of biological functions, canonical pathways, and genetic networks linked to IMF-related phenotypes. In summary, our GWAS and network predictions are supported by the current literature and suggest a cooperative role for the 3 TF and other interacting genes including CAPN6, STC2, MAP2K4, EYA1, COPS5, XKR4, NR2E1, TOX, ATF1, ASPH, TGS1, and TTPA as modulators of carcass and meat quality traits in beef cattle.
Collapse
Affiliation(s)
- Y Ramayo-Caldas
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia Departament de Ciencia Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain INRA, UMR1313 Génétique Animale et Biologie Intégrative (GABI), Domaine de Vilvert, Bâtiment GABI-320, 78352 Jouy-en-Josas, France
| | - M R S Fortes
- The University of Queensland, Queensland Alliance for Agriculture and Food Innovation, Center for Animal Science, QLD 4062, Australia
| | - N J Hudson
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia
| | - L R Porto-Neto
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia
| | - S Bolormaa
- Victorian Department of Environment and Primary Industries, Bundoora, VIC 3083, Australia
| | - W Barendse
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia
| | - M Kelly
- The University of Queensland, Queensland Alliance for Agriculture and Food Innovation, Center for Animal Science, QLD 4062, Australia
| | - S S Moore
- The University of Queensland, Queensland Alliance for Agriculture and Food Innovation, Center for Animal Science, QLD 4062, Australia
| | - M E Goddard
- Victorian Department of Environment and Primary Industries, Bundoora, VIC 3083, Australia School of Land and Environment, University of Melbourne, Parkville, VIC 3010, Australia
| | - S A Lehnert
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia
| | - A Reverter
- CSIRO Food Futures Flagship and CSIRO Animal, Food and Health Sciences, 306 Carmody Road, St. Lucia, Brisbane, QLD 4067, Australia
| |
Collapse
|
25
|
Felthaus O, Gosau M, Morsczeck C. ZBTB16 induces osteogenic differentiation marker genes in dental follicle cells independent from RUNX2. J Periodontol 2013; 85:e144-51. [PMID: 24359167 DOI: 10.1902/jop.2013.130445] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Dental follicle cells (DFCs) are neural crest cell-derived cells and the genuine precursor cells of cementoblast and alveolar osteoblasts. After osteogenic differentiation, expression levels of the transcription factor zinc factor and BTB domain containing 16 (ZBTB16) were significantly increased. ZBTB16 is associated with the process of osteogenic differentiation in bone marrow-derived mesenchymal stem cells and crucial for the expression of the osteogenic transcription factor runt-related transcription factor 2 (RUNX2). It is proposed that ZBTB16 plays also a crucial role for the differentiation of DFCs into osteoblasts. METHODS In this study, the differentiation of DFCs by alkaline phosphatase (ALP) activity measurement, alizarin red staining, and electron-dispersive x-ray spectrometry (EDX) analysis is investigated. The expression of ZBTB16 during osteogenic differentiation and the expression of osteogenic differentiation markers were assessed by real-time reverse transcription polymerase chain reaction. Glucocorticoid stimulation was inhibited using RU486 (11β-[p-(Dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one), and ZBTB16 was overexpressed via transient transfection of an expression vector. RESULTS After the initiation of osteogenic differentiation, ZBTB16 levels were increased highly in DFCs, whereas RUNX2 was expressed constitutively only. An EDX analysis verified the differentiation of DFCs into osteoblast-like cells because clusters of mineralization consisted of hydroxyapatite. ZBTB16 induced the expression of nuclear receptor subfamily 4, group A, member 3; osteocalcin; and stanniocalcin 1 (STC1) but not of RUNX2 and ALP in DFCs. STC1 was upregulated in DFCs downstream of ZBTB16 and after the osteogenic differentiation. The overexpression of STC1 in DFCs increased the expression of ZBTB16 and specific markers for biomineralization. CONCLUSIONS The present study shows that ZBTB16 induced the expression of osteogenic differentiation markers independently of RUNX2. Moreover, STC1 is a new candidate for the evaluation of late mechanisms of osteogenic differentiation downstream of ZBTB16.
Collapse
Affiliation(s)
- Oliver Felthaus
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
26
|
Satija NK, Sharma D, Afrin F, Tripathi RP, Gangenahalli G. High throughput transcriptome profiling of lithium stimulated human mesenchymal stem cells reveals priming towards osteoblastic lineage. PLoS One 2013; 8:e55769. [PMID: 23383279 PMCID: PMC3559497 DOI: 10.1371/journal.pone.0055769] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 01/04/2013] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) present in the bone marrow are the precursors of osteoblasts, chondrocytes and adipocytes, and hold tremendous potential for osteoregenerative therapy. However, achieving directed differentiation into osteoblasts has been a major concern. The use of lithium for enhancing osteogenic differentiation has been documented in animal models but its effect in humans is not clear. We, therefore, performed high throughput transcriptome analysis of lithium-treated hMSCs to identify altered gene expression and its relevance to osteogenic differentiation. Our results show suppression of proliferation and enhancement of alkaline phosphatase (ALP) activity upon lithium treatment of hMSCs under non-osteogenic conditions. Microarray profiling of lithium-stimulated hMSC revealed decreased expression of adipogenic genes (CEBPA, CMKLR1, HSD11B1) and genes involved in lipid biosynthesis. Interestingly, osteoclastogenic factors and immune responsive genes (IL7, IL8, CXCL1, CXCL12, CCL20) were also downregulated. Negative transcriptional regulators of the osteogenic program (TWIST1 and PBX1) were suppressed while genes involved in mineralization like CLEC3B and ATF4 were induced. Gene ontology analysis revealed enrichment of upregulated genes related to mesenchymal cell differentiation and signal transduction. Lithium priming led to enhanced collagen 1 synthesis and osteogenic induction of lithium pretreated MSCs resulted in enhanced expression of Runx2, ALP and bone sialoprotein. However, siRNA-mediated knockdown of RRAD, CLEC3B and ATF4 attenuated lithium-induced osteogenic priming, identifying a role for RRAD, a member of small GTP binding protein family, in osteoblast differentiation. In conclusion, our data highlight the transcriptome reprogramming potential of lithium resulting in higher propensity of lithium "primed" MSCs for osteoblastic differentiation.
Collapse
Affiliation(s)
- Neeraj Kumar Satija
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Deepa Sharma
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Farhat Afrin
- Department of Biotechnology, Hamdard University, Hamdard Nagar, New Delhi, India
| | - Rajendra P. Tripathi
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Gurudutta Gangenahalli
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
27
|
Gosau M, Götz W, Felthaus O, Ettl T, Jäger A, Morsczeck C. Comparison of the differentiation potential of neural crest derived progenitor cells from apical papilla (dNC-PCs) and stem cells from exfoliated deciduous teeth (SHED) into mineralising cells. Arch Oral Biol 2012; 58:699-706. [PMID: 23261253 DOI: 10.1016/j.archoralbio.2012.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 11/05/2012] [Accepted: 11/07/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Recently, cells from the apical papilla of retained human third molars (dental neural crest-derived progenitor cells, dNC-PCs) have been isolated and characterised as multipotent progenitor cells. Nonetheless, molecular processes during differentiation into mineralising cells are still unknown. This study evaluated the osteogenic/odontogenic differentiation of dNC-PCs under in vitro conditions and compared these cells with already known odontoblast precursor cells (dental stem cells from exfoliated human deciduous teeth, SHED). METHODS The differentiation of dNC-PCs and SHED under in vitro conditions was verified by Alizarin red staining (mineralisation), alkaline phosphatase activity and the expression of osteogenic/odontogenic markers (RT-PCRs). The genome wide expression-profiles were investigated with Affymetrix DNA-microarrays and the cell migration with a gel spot cell migration assay. RESULTS In our study dNC-PCs differentiated like SHED in mineralising cells. The expression of odontoblast markers suggested that dNC-PCs and SHED differentiated into different types of odontoblasts. This supposition was supported by genome wide gene expression profiles of dNC-PCs and SHED after cell differentiation. Typical biological processes of undifferentiated cells, for example "mitosis", were regulated in dNC-PCs. In SHED biological processes like "response to wounding" or "cell migration" were regulated, which are associated with replacement odontoblasts and their precursors. Moreover, a gel-spot assay revealed that SHED migrated faster than dNC-PCs. CONCLUSION Our results suggest that dNC-PCs are precursors for primary odontoblasts, whereas SHED differentiate into replacement odontoblasts. These different odontogenic differentiation potentials of dNC-PCs and SHED have to be considered for cellular therapies and tissue engineering approaches in the future.
Collapse
Affiliation(s)
- Martin Gosau
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
28
|
Mid-gestational gene expression profile in placenta and link to pregnancy complications. PLoS One 2012; 7:e49248. [PMID: 23145134 PMCID: PMC3492272 DOI: 10.1371/journal.pone.0049248] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 10/04/2012] [Indexed: 12/25/2022] Open
Abstract
Despite the importance of placenta in mediating rapid physiological changes in pregnancy, data on temporal dynamics of placental gene expression are limited. We completed the first transcriptome profiling of human placental gene expression dynamics (GeneChips, Affymetrix®; ∼47,000 transcripts) from early to mid-gestation (n = 10; gestational weeks 5–18) and report 154 genes with significant transcriptional changes (ANOVA, FDR P<0.1). TaqMan RT-qPCR analysis (n = 43; gestational weeks 5–41) confirmed a significant (ANOVA and t-test, FDR P<0.05) mid-gestational peak of placental gene expression for BMP5, CCNG2, CDH11, FST, GATM, GPR183, ITGBL1, PLAGL1, SLC16A10 and STC1, followed by sharp decrease in mRNA levels at term (t-test, FDR P<0.05). We hypothesized that normal course of late pregnancy may be affected when genes characteristic to mid-gestation placenta remain highly expressed until term, and analyzed their expression in term placentas from normal and complicated pregnancies [preeclampsia (PE), n = 12; gestational diabetes mellitus (GDM), n = 12; small- and large-for-gestational-age newborns (SGA, LGA), n = 12+12]. STC1 (stanniocalcin 1) exhibited increased mRNA levels in all studied complications, with the most significant effect in PE- and SGA-groups (t-test, FDR P<0.05). In post-partum maternal plasma, the highest STC1 hormone levels (ELISA, n = 129) were found in women who had developed PE and delivered a SGA newborn (median 731 vs 418 pg/ml in controls; ANCOVA, P = 0.00048). Significantly higher expression (t-test, FDR P<0.05) of CCNG2 and LYPD6 accompanied with enhanced immunostaining of the protein was detected in placental sections of PE and GDM cases (n = 15). Our study demonstrates the importance of temporal dynamics of placental transcriptional regulation across three trimesters of gestation. Interestingly, many genes with high expression in mid-gestation placenta have also been implicated in adult complex disease, promoting the discussion on the role of placenta in developmental programming. The discovery of elevated maternal plasma STC1 in pregnancy complications warrants further investigations of its potential as a biomarker.
Collapse
|
29
|
Lui JC, Nilsson O, Chan Y, Palmer CD, Andrade AC, Hirschhorn JN, Baron J. Synthesizing genome-wide association studies and expression microarray reveals novel genes that act in the human growth plate to modulate height. Hum Mol Genet 2012; 21:5193-201. [PMID: 22914739 DOI: 10.1093/hmg/dds347] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Previous meta-analysis of genome-wide association (GWA) studies has identified 180 loci that influence adult height. However, each GWA locus typically comprises a set of contiguous genes, only one of which presumably modulates height. We reasoned that many of the causative genes within these loci influence height because they are expressed in and function in the growth plate, a cartilaginous structure that causes bone elongation and thus determines stature. Therefore, we used expression microarray studies of mouse and rat growth plate, human disease databases and a mouse knockout phenotype database to identify genes within the GWAS loci that are likely required for normal growth plate function. Each of these approaches identified significantly more genes within the GWA height loci than at random genomic locations (P < 0.0001 each), supporting the validity of the approach. The combined analysis strongly implicates 78 genes in growth plate function, including multiple genes that participate in PTHrP-IHH, BMP and CNP signaling, and many genes that have not previously been implicated in the growth plate. Thus, this analysis reveals a large number of novel genes that regulate human growth plate chondrogenesis and thereby contribute to the normal variations in human adult height. The analytic approach developed for this study may be applied to GWA studies for other common polygenic traits and diseases, thus providing a new general strategy to identify causative genes within GWA loci and to translate genetic associations into mechanistic biological insights.
Collapse
Affiliation(s)
- Julian C Lui
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Yeung BHY, Law AYS, Wong CKC. Evolution and roles of stanniocalcin. Mol Cell Endocrinol 2012; 349:272-80. [PMID: 22115958 DOI: 10.1016/j.mce.2011.11.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/07/2011] [Indexed: 12/11/2022]
Abstract
In fish, stanniocalcin-1 (STC1) is a key endocrine factor that acts on gill, intestine and kidney to regulate serum calcium and phosphate homeostasis. The recent identification and study of mammalian STCs (STC1 and STC2) revealed that the hormones are made in virtually all tissues and they act primarily as paracrine/autocrine factors to regulate various biological functions. Based on their ubiquitous expression patterns and generally undetectable levels in blood serum, it is unlikely that the mammalian STCs play important roles in serum Ca(2+)/P(i) homeostasis. However current evidences still support the local action of STCs in Ca(2+) and P(i) transport, probably via their action on Ca(2+)-channels and Na(+)/P(i) co-transporter. At present, information about the sequence, expression and distribution of the STC receptor(s) is lacking. However, recent emerging evidence hints the involvement of STC1 and STC2 in the sub-cellular functions of mitochondria and endoplasmic reticulum respectively, particularly responding to oxidative stress and unfolded protein response. With increasing evidence that demonstrates the local actions of STCs, the focus of the research has been moved to cellular inflammation and carcinogenesis. This review integrates the information available on STCs in fish and mammals, focusing mainly on their embryonic origin, tissue distribution, their potential regulatory mechanisms and the modes of action, and their physiological and pathophysiological functions, particularly in cancer biology.
Collapse
Affiliation(s)
- B H Y Yeung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | | | |
Collapse
|
31
|
Richards TDJ, Fenton AL, Syed R, Wagner GF. Characterization of stanniocalcin-1 receptors in the rainbow trout. ISRN ENDOCRINOLOGY 2012; 2012:257841. [PMID: 22474594 PMCID: PMC3302014 DOI: 10.5402/2012/257841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 09/20/2011] [Indexed: 01/08/2023]
Abstract
Mammalian stanniocalcin-1 (STC-1) is one of several ligands targeted to mitochondria. High affinity STC-1 receptors are present on the mitochondrial membranes of nephron cells, myocytes, and hepatocytes, to enable ligand sequestration within the matrix. However, STC-1 receptors have not been characterized in fish. Nor is it known if mitochondrial targeting occurs in fish. The aim of the study was to address these questions. Saturation binding assays were carried out to obtain estimates of KD
and
Bmax. They revealed the presence of saturable, high-affinity receptors on both membranes and mitochondria of liver, muscle, and gill filament. In situ ligand binding (ISLB) was used to localize receptors at the histological level and revealed some unexpected findings. In cranium, for instance, receptors were found mainly in the cartilage matrix, as opposed to the chondrocytes. In brain, the majority of receptors were located on neuropil areas as opposed to neuronal cell bodies. In skeletal muscle, receptors were confined to periodic striations, tentatively identified as the Z lines. Receptors were even found on STC-1 producing corpuscles of Stannius cells, raising the possibility of there being an autocrine feedback loop or, perhaps, a soluble binding protein that is released with the ligand to regulate its bioavailability.
Collapse
Affiliation(s)
- Timothy D J Richards
- Department of Physiology and Pharmacology, Faculty of Medicine and Dentistry, University of Western Ontario, London, ON, Canada N6A 5C1
| | | | | | | |
Collapse
|
32
|
Khoshniat S, Bourgine A, Julien M, Weiss P, Guicheux J, Beck L. The emergence of phosphate as a specific signaling molecule in bone and other cell types in mammals. Cell Mol Life Sci 2011; 68:205-18. [PMID: 20848155 PMCID: PMC11114507 DOI: 10.1007/s00018-010-0527-z] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 08/02/2010] [Accepted: 08/31/2010] [Indexed: 02/07/2023]
Abstract
Although considerable advances in our understanding of the mechanisms of phosphate homeostasis and skeleton mineralization have recently been made, little is known about the initial events involving the detection of changes in the phosphate serum concentrations and the subsequent downstream regulation cascade. Recent data has strengthened a long-established hypothesis that a phosphate-sensing mechanism may be present in various organs. Such a phosphate sensor would detect changes in serum or local phosphate concentration and would inform the body, the local environment, or the individual cell. This suggests that phosphate in itself could represent a signal regulating multiple factors necessary for diverse biological processes such as bone or vascular calcification. This review summarizes findings supporting the possibility that phosphate represents a signaling molecule, particularly in bone and cartilage, but also in other tissues. The involvement of various signaling pathways (ERK1/2), transcription factors (Fra-1, Runx2) and phosphate transporters (PiT1, PiT2) is discussed.
Collapse
Affiliation(s)
- Solmaz Khoshniat
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Annabelle Bourgine
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Marion Julien
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Pierre Weiss
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Jérôme Guicheux
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Laurent Beck
- Growth and Signalling Research Center, INSERM, U845, 75015 Paris, France
- Faculté de Médecine, Centre de Recherche, INSERM U845, Université Paris Descartes, 156 Rue de Vaugirard, 75015 Paris, France
| |
Collapse
|
33
|
dos Santos MT, Trindade DM, Gonçalves KDA, Bressan GC, Anastassopoulos F, Yunes JA, Kobarg J. Human stanniocalcin-1 interacts with nuclear and cytoplasmic proteins and acts as a SUMO E3 ligase. MOLECULAR BIOSYSTEMS 2010; 7:180-93. [PMID: 21042649 DOI: 10.1039/c0mb00088d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human stanniocalcin-1 (STC1) is a glycoprotein that has been implicated in different physiological process, including angiogenesis, apoptosis and carcinogenesis. Here we identified STC1 as a putative molecular marker for the leukemic bone marrow microenvironment and identified new interacting protein partners for STC1. Seven selected interactions retrieved from yeast two-hybrid screens were confirmed by GST-pull down assays in vitro. The N-terminal region was mapped to be the region that mediates the interaction with cytoplasmic, mitochondrial and nuclear proteins. STC1 interacts with SUMO-1 and several proteins that have been shown to be SUMOylated and localized to SUMOylation related nuclear bodies. Although STC1 interacts with SUMO-1 and has a high theoretical prediction score for a SUMOylation site, endogenous co-immunoprecipitation and in vitro SUMOylation assays with the purified recombinant protein could not detect STC1 SUMOylation. However, when we tested STC1 for SUMO E3 ligase activity, we found in an in vitro assay, that it significantly increases the SUMOylation of two other proteins. Confocal microscopic subcellular localization studies using both transfected cells and specific antibodies for endogenous STC1 revealed a cytoplasmic and nuclear deposition, the latter in the form of some specific dot-like substructure resembling SUMOylation related nuclear bodies. Together, these findings suggest a new role for STC1 in SUMOylation pathways, in nuclear bodies.
Collapse
Affiliation(s)
- Marcos Tadeu dos Santos
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Rua Giuseppe Máximo Scolfaro 10.000, CP6192, 13084-971 Campinas, SP, Brasil.
| | | | | | | | | | | | | |
Collapse
|