1
|
Alahdad N, Hamidpour SK, Yazdanpanah MA, Amiri M, Alizadeh R, Rezayat SM, Tavakol S. Nitric oxide synthases: A delicate dance between bone regeneration and neuronal birth. Biomed Pharmacother 2025; 187:118105. [PMID: 40294491 DOI: 10.1016/j.biopha.2025.118105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/23/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025] Open
Abstract
Spinal cord injury (SCI) is a devastating condition resulting from traumatic or nontraumatic injury/chronic disorder. The pathogenesis of SCI necessitates a comprehensive approach, as it involves therapeutic strategies addressing both bone (spine) and neural (spinal cord) damage. This review centers on the pivotal role of nitric oxide (NO) and its synthesizing enzymes, nitric oxide synthases (NOS), in mediating the crosstalk between osteogenesis and neurogenesis. NO's effects are context-dependent, exhibiting a delicate balance between beneficial and detrimental actions. Reduced levels of nitric oxide (NO), primarily derived from endothelial NOS (eNOS), tipically stimulate osteoblast activity and promote neurogenesis by influencing neural stem cell (NSC) migration and differentiation. Conversely, elevated NO levels, predominantly from inducible NOS (iNOS), tipically triggered by inflammation, inhibit both processes through pro-apoptotic mechanisms. Nevertheless, these phenomena are not merely simplistic; they can be influenced by a variety of other factors. We explore the intricate interplay of NO/NOS with key signaling pathways crucial in neurogenesis and osteogenesis, including mechanical stimuli, Wnt, interleukins, BMPs, NF-κB, etc., revealing their influence on neuroinflammation, neurogenesis, and osteoblast differentiation. The temporal and spatial dynamics of NO/NOS activity and the implications for therapeutic intervention have been discussed. Precise modulation of NO levels and NOS isoforms, potentially through targeted therapies manipulating these interacting signaling pathways, emerges as a promising strategy for promoting bone and neural regeneration. This review highlights the critical need for a balanced approach in therapeutic strategies to harness the beneficial effects of NO/NOS while mitigating its detrimental consequences.
Collapse
Affiliation(s)
- Niloofar Alahdad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Shayesteh Kokabi Hamidpour
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Mohammad Ali Yazdanpanah
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Mobina Amiri
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Research and Development, Tavakol BioMimetic Technologies Company, Tehran, Iran.
| |
Collapse
|
2
|
Juraver-Geslin H, Devotta A, Saint-Jeannet JP. Developmental roles of natriuretic peptides and their receptors. Cells Dev 2023; 176:203878. [PMID: 37742795 PMCID: PMC10841480 DOI: 10.1016/j.cdev.2023.203878] [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: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Natriuretic peptides and their receptors are implicated in the physiological control of blood pressure, bone growth, and cardiovascular and renal homeostasis. They mediate their action through the modulation of intracellular levels of cGMP and cAMP, two second-messengers that have broad biological roles. In this review, we briefly describe the major players of this signaling pathway and their physiological roles in the adult, and discuss several reports describing their activity in the control of various aspects of embryonic development in several species. While the core components of this signaling pathway are well conserved, their functions have diverged in the embryo and the adult to control a diverse array of biological processes.
Collapse
Affiliation(s)
- Hugo Juraver-Geslin
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Arun Devotta
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA.
| |
Collapse
|
3
|
Feibel D, Kwiatkowski A, Opländer C, Grieb G, Windolf J, Suschek CV. Enrichment of Bone Tissue with Antibacterially Effective Amounts of Nitric Oxide Derivatives by Treatment with Dielectric Barrier Discharge Plasmas Optimized for Nitrogen Oxide Chemistry. Biomedicines 2023; 11:biomedicines11020244. [PMID: 36830781 PMCID: PMC9953554 DOI: 10.3390/biomedicines11020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
Cold atmospheric plasmas (CAPs) generated by dielectric barrier discharge (DBD), particularly those containing higher amounts of nitric oxide (NO) or NO derivates (NOD), are attracting increasing interest in medical fields. In the present study, we, for the first time, evaluated DBD-CAP-induced NOD accumulation and therapeutically relevant NO release in calcified bone tissue. This knowledge is of great importance for the development of new therapies against bacterial-infectious complications during bone healing, such as osteitis or osteomyelitis. We found that by modulating the power dissipation in the discharge, it is possible (1) to significantly increase the uptake of NODs in bone tissue, even into deeper regions, (2) to significantly decrease the pH in CAP-exposed bone tissue, (3) to induce a long-lasting and modulable NO production in the bone samples as well as (4) to significantly protect the treated bone tissue against bacterial contaminations, and to induce a strong bactericidal effect in bacterially infected bone samples. Our results strongly suggest that the current DBD technology opens up effective NO-based therapy options in the treatment of local bacterial infections of the bone tissue through the possibility of a targeted modulation of the NOD content in the generated CAPs.
Collapse
Affiliation(s)
- Dennis Feibel
- Department for Orthopedics and Trauma Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Alexander Kwiatkowski
- Department for Orthopedics and Trauma Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christian Opländer
- Institute for Research in Operative Medicine (IFOM), Cologne-Merheim Medical Center, University Witten/Herdecke, 58455 Witten-Herdecke, Germany
| | - Gerrit Grieb
- Department of Plastic Surgery and Hand Surgery, Burn Centre, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Joachim Windolf
- Department for Orthopedics and Trauma Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christoph V. Suschek
- Department for Orthopedics and Trauma Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
- Correspondence:
| |
Collapse
|
4
|
Wagner BM, Robinson JW, Prickett TCR, Espiner EA, Khosla S, Gaddy D, Suva LJ, Potter LR. Guanylyl Cyclase-B Dependent Bone Formation in Mice is Associated with Youth, Increased Osteoblasts, and Decreased Osteoclasts. Calcif Tissue Int 2022; 111:506-518. [PMID: 35947145 DOI: 10.1007/s00223-022-01014-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/15/2022] [Indexed: 11/02/2022]
Abstract
C-type natriuretic peptide (CNP) activation of guanylyl cyclase-B (GC-B) catalyzes the synthesis of cGMP in chondrocytes and osteoblasts. Elevated cGMP stimulates long bone growth, and inactivating mutations in CNP or GC-B reduce cGMP, which causes dwarfism. GC-B7E/7E mice that express a GC-B mutant that cannot be inactivated by dephosphorylation exhibit increased CNP-dependent GC-B activity, which increases bone length, as well as bone mass and strength. Importantly, how GC-B increases bone mass is not known. Here, we injected 12-week-old, wild type mice once daily for 28 days with or without BMN-111 (Vosoritide), a proteolytically resistant CNP analog. We found that BMN-111 treated mice had elevated levels of osteocalcin and collagen 1 C-terminal telopeptide (CTX) as well as increased osteoblasts and osteoclasts. In BMN-111 injected mice, tibial mRNAs for Rank ligand and osteoprotegrin were increased and decreased, respectively, whereas sclerostin mRNA was elevated 400-fold, consistent with increased osteoclast activity and decreased osteoblast activity. Mineral apposition rates and trabecular bone mass were not elevated in response to BMN-111. Because 9-week-old male GC-B7E/7E mice have increased bone mass but do not exhibit increased mineral apposition rates, we examined 4-week-old male GC-B7E/7E mice and found that these animals had increased serum osteocalcin, but not CTX. Importantly, tibias from these mice had 37% more osteoblasts, 26% fewer osteoclasts as well as 36% and 40% higher mineral apposition and bone formation rates, respectively. We conclude that GC-B-dependent bone formation is coupled to an early juvenile process that requires both increased osteoblasts and decreased osteoclasts.
Collapse
Affiliation(s)
- Brandon M Wagner
- Departments of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jerid W Robinson
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN, USA
| | | | - Eric A Espiner
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dana Gaddy
- Departments of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Larry J Suva
- Departments of Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Lincoln R Potter
- Departments of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Molecular Mechanism of Induction of Bone Growth by the C-Type Natriuretic Peptide. Int J Mol Sci 2022; 23:ijms23115916. [PMID: 35682595 PMCID: PMC9180634 DOI: 10.3390/ijms23115916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 12/10/2022] Open
Abstract
The skeletal development process in the body occurs through sequential cellular and molecular processes called endochondral ossification. Endochondral ossification occurs in the growth plate where chondrocytes differentiate from resting, proliferative, hypertrophic to calcified zones. Natriuretic peptides (NPTs) are peptide hormones with multiple functions, including regulation of blood pressure, water-mineral balance, and many metabolic processes. NPTs secreted from the heart activate different tissues and organs, working in a paracrine or autocrine manner. One of the natriuretic peptides, C-type natriuretic peptide-, induces bone growth through several mechanisms. This review will summarize the knowledge, including the newest discoveries, of the mechanism of CNP activation in bone growth.
Collapse
|
6
|
Marchini M, Ashkin MR, Bellini M, Sun MMG, Workentine ML, Okuyan HM, Krawetz R, Beier F, Rolian C. A Na +/K + ATPase Pump Regulates Chondrocyte Differentiation and Bone Length Variation in Mice. Front Cell Dev Biol 2022; 9:708384. [PMID: 34970538 PMCID: PMC8712571 DOI: 10.3389/fcell.2021.708384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022] Open
Abstract
The genetic and developmental mechanisms involved in limb formation are relatively well documented, but how these mechanisms are modulated by changes in chondrocyte physiology to produce differences in limb bone length remains unclear. Here, we used high throughput RNA sequencing (RNAseq) to probe the developmental genetic basis of variation in limb bone length in Longshanks, a mouse model of experimental evolution. We find that increased tibia length in Longshanks is associated with altered expression of a few key endochondral ossification genes such as Npr3, Dlk1, Sox9, and Sfrp1, as well reduced expression of Fxyd2, a facultative subunit of the cell membrane-bound Na+/K+ ATPase pump (NKA). Next, using murine tibia and cell cultures, we show a dynamic role for NKA in chondrocyte differentiation and in bone length regulation. Specifically, we show that pharmacological inhibition of NKA disrupts chondrocyte differentiation, by upregulating expression of mesenchymal stem cell markers (Prrx1, Serpina3n), downregulation of chondrogenesis marker Sox9, and altered expression of extracellular matrix genes (e.g., collagens) associated with proliferative and hypertrophic chondrocytes. Together, Longshanks and in vitro data suggest a broader developmental and evolutionary role of NKA in regulating limb length diversity.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Mitchell R Ashkin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Melina Bellini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew Lloyd Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hamza Malik Okuyan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Roman Krawetz
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Feil R, Lehners M, Stehle D, Feil S. Visualising and understanding cGMP signals in the cardiovascular system. Br J Pharmacol 2021; 179:2394-2412. [PMID: 33880767 DOI: 10.1111/bph.15500] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/14/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022] Open
Abstract
cGMP is an important signalling molecule in humans. Fluorescent cGMP biosensors have emerged as powerful tools for the sensitive analysis of cGMP pathways at the single-cell level. Here, we briefly outline cGMP's multifaceted role in (patho)physiology and pharmacotherapy. Then we summarise what new insights cGMP imaging has provided into endogenous cGMP signalling and drug action, with a focus on the cardiovascular system. Indeed, the use of cGMP biosensors has led to several conceptual advances, such as the discovery of local, intercellular and mechanosensitive cGMP signals. Importantly, single-cell imaging can provide valuable information about the heterogeneity of cGMP signals within and between individual cells of an isolated cell population or tissue. We also discuss current challenges and future directions of cGMP imaging, such as the direct visualisation of cGMP microdomains, simultaneous monitoring of cGMP and other signalling molecules and, ultimately, cGMP imaging in tissues and animals under close-to-native conditions.
Collapse
Affiliation(s)
- Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Anastasio AT, Paniagua A, Diamond C, Ferlauto HR, Fernandez-Moure JS. Nanomaterial Nitric Oxide Delivery in Traumatic Orthopedic Regenerative Medicine. Front Bioeng Biotechnol 2021; 8:592008. [PMID: 33537289 PMCID: PMC7849904 DOI: 10.3389/fbioe.2020.592008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
Achieving bone fracture union after trauma represents a major challenge for the orthopedic surgeon. Fracture non-healing has a multifactorial etiology and there are many risk factors for non-fusion. Environmental factors such as wound contamination, infection, and open fractures can contribute to non-healing, as can patient specific factors such as poor vascular status and improper immunologic response to fracture. Nitric oxide (NO) is a small, neutral, hydrophobic, highly reactive free radical that can diffuse across local cell membranes and exert paracrine functions in the vascular wall. This molecule plays a role in many biologic pathways, and participates in wound healing through decontamination, mediating inflammation, angiogenesis, and tissue remodeling. Additionally, NO is thought to play a role in fighting wound infection by mitigating growth of both Gram negative and Gram positive pathogens. Herein, we discuss recent developments in NO delivery mechanisms and potential implications for patients with bone fractures. NO donors are functional groups that store and release NO, independent of the enzymatic actions of NOS. Donor molecules include organic nitrates/nitrites, metal-NO complexes, and low molecular weight NO donors such as NONOates. Numerous advancements have also been made in developing mechanisms for localized nanomaterial delivery of nitric oxide to bone. NO-releasing aerogels, sol- gel derived nanomaterials, dendrimers, NO-releasing micelles, and core cross linked star (CCS) polymers are all discussed as potential avenues of NO delivery to bone. As a further target for improved fracture healing, 3d bone scaffolds have been developed to include potential for nanoparticulated NO release. These advancements are discussed in detail, and their potential therapeutic advantages are explored. This review aims to provide valuable insight for translational researchers who wish to improve the armamentarium of the feature trauma surgeon through use of NO mediated augmentation of bone healing.
Collapse
Affiliation(s)
| | - Ariana Paniagua
- Duke University School of Medicine, Durham, NC, United States
| | - Carrie Diamond
- Duke University School of Medicine, Durham, NC, United States
| | | | | |
Collapse
|
9
|
Hanley PC, Kanwar HS, Martineau C, Levine MA. Short Stature is Progressive in Patients with Heterozygous NPR2 Mutations. J Clin Endocrinol Metab 2020; 105:5877389. [PMID: 32720985 PMCID: PMC7442278 DOI: 10.1210/clinem/dgaa491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/22/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND NPR2 encodes atrial natriuretic peptide receptor B (ANPRB), a regulator of skeletal growth. Biallelic loss-of-function mutations in NPR2 result in acromesomelic dysplasia Maroteaux type (AMDM; OMIM 602875), while heterozygous mutations may account for 2% to 6% of idiopathic short stature (ISS). OBJECTIVE Describe the physical proportions and growth characteristics of an extended family with novel NPR2 mutations including members with AMDM, ISS, or normal stature. DESIGN AND PARTICIPANTS We performed whole exome sequencing in 2 healthy parents and 2 children with AMDM. Detailed genotyping and phenotyping were performed on members of a multigenerational family in an academic medical center. We expressed mutant proteins in mammalian cells and characterized expression and function. RESULTS The sisters with AMDM were compound heterozygotes for missense mutations in the NPR2 gene, a novel p.P93S (maternal) and the previously reported p.R989L (paternal). Both mutant ANPRB proteins were normally expressed in HEK293T cells and exhibited dominant negative effects on wild-type ANPRB catalytic activity. Heterozygous relatives had proportionate short stature (height z-scores -2.06 ± 0.97, median ± SD) compared with their wild-type siblings (-1.37 ± 0.59). Height z-scores progressively and significantly decreased as NPR2-heterozygous children matured, while remaining constant in their wild-type siblings. CONCLUSIONS Biallelic NPR2 mutations cause severe skeletal dysplasia (AMDM), whereas heterozygous mutations lead to a subtler phenotype characterized by progressive short stature with by increasing loss of height potential with age.
Collapse
Affiliation(s)
- Patrick C Hanley
- Division of Endocrinology, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware
- Correspondence and Reprint Requests: Patrick C. Hanley, MD, Division of Endocrinology, Nemours Alfred I. DuPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE 19803. E-mail:
| | - Harsh S Kanwar
- Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Corine Martineau
- Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Michael A Levine
- Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
A novel nonsense mutation in NPR2 gene causing Acromesomelic dysplasia, type Maroteaux in a consanguineous family in Southern Punjab (Pakistan). Genes Genomics 2020; 42:847-854. [PMID: 32506268 PMCID: PMC7374443 DOI: 10.1007/s13258-020-00955-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/12/2020] [Indexed: 11/16/2022]
Abstract
Background Acromesomelic dysplasia, type Maroteaux (AMDM) is a rare skeletal dysplasia following autosomal recessive mode of inheritance and characterized by abnormal growth plates, short and abnormal bones in the extremities and spine. Objective Present study was designed to report the molecular basis of AMDM in enrolled consanguineous family from Pakistan. Methods A consanguineous family from Vehari District in Pakistan having multiple siblings suffering from AMDM was enrolled in present study. Whole exome sequencing (WES) approach was adopted to identify causative agent of AMDM. Human full length NPR2 gene and sequence with nonsense mutation was amplified by using Myc-tagged pXN vector and transformed in E. coli DH5α cells to confirm mutation. SDS-PAGE and Western blotting were done to confirm the production of truncated protein. Computational three dimensional structure generation through homology modeling approach was done to compare protein structure between patients and controls. Results WES reveled a nonsense mutation (c.613 C>T, p.R205X) in exon 1 of NPR2 gene leading to premature termination codon in mRNA of NPR2 gene resulting in a truncated protein with 204 amino acid residues that was confirmed by SDS-PAGE and Western blotting. Sanger sequencing confirmed that mutation in all subjects and mutation followed Mendalian pattern of inheritance. Multiple sequence alignment by ClustalW revealed that mutated domain of NPR2 is conserved region. Proetin structure comparison revealed a significant structural part of NPR2 was missing in truncated protein as compared to control. Conclusion We are reporting that a novel nonsense mutation (c.613 C>T, p.R205X) in exon 1 of NPR2 gene is causing AMDM in a consanguineous Pakistani family.
Collapse
|
11
|
Robinson JW, Blixt NC, Norton A, Mansky KC, Ye Z, Aparicio C, Wagner BM, Benton AM, Warren GL, Khosla S, Gaddy D, Suva LJ, Potter LR. Male mice with elevated C-type natriuretic peptide-dependent guanylyl cyclase-B activity have increased osteoblasts, bone mass and bone strength. Bone 2020; 135:115320. [PMID: 32179168 DOI: 10.1016/j.bone.2020.115320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/27/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
C-type natriuretic peptide (CNP) activation of guanylyl cyclase (GC)-B, also known as NPR2, stimulates cGMP synthesis and bone elongation. CNP activation requires the phosphorylation of multiple GC-B residues and dephosphorylation inactivates the receptor. GC-B7E/7E knockin mice, expressing a glutamate-substituted, "pseudophosphorylated," form of GC-B, exhibit increased CNP-dependent GC activity. Since mutations that constitutively activate GC-B in the absence of CNP result in low bone mineral density in humans, we determined the skeletal phenotype of 9-week old male GC-B7E/7E mice. Unexpectedly, GC-B7E/7E mice have significantly greater tibial and L5 vertebral trabecular bone volume fraction, tibial trabecular number, and tibial bone mineral density. Cortical cross-sectional area, cortical thickness, periosteal diameter and cortical cross-sectional moment of inertia were also significantly increased in GC-B7E/7E tibiae. Three-point bending measurements demonstrated that the mutant tibias and femurs had greater ultimate load, stiffness, energy to ultimate load, and energy to failure. No differences in microhardness indicated similar bone quality at the tissue level between the mutant and wildtype bones. Procollagen 1 N-terminal propeptide and osteocalcin were elevated in serum, and osteoblast number per bone perimeter and osteoid width per bone perimeter were elevated in tibias from the mutant mice. In contrast to mutations that constitutively activate GC-B, we report that mutations that enhance GC-B activity only in the presence of its natural ligand, increase bone mass, bone strength, and the number of active osteoblasts at the bone surface.
Collapse
Affiliation(s)
- Jerid W Robinson
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas C Blixt
- Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Andrew Norton
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Kim C Mansky
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Zhou Ye
- Restorative Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Conrado Aparicio
- Restorative Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Brandon M Wagner
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Andrew M Benton
- Department of Physical Therapy, Georgia State University, Atlanta, GA, USA
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, GA, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Larry J Suva
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Lincoln R Potter
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA; Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
12
|
An SY, Lee HJ, Lee SC, Heo JS. Supplement of nitric oxide through calcium carbonate-based nanoparticles contributes osteogenic differentiation of mouse embryonic stem cells. Tissue Cell 2020; 66:101390. [PMID: 32933713 DOI: 10.1016/j.tice.2020.101390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 12/16/2022]
Abstract
This study investigated the delivery of S-nitrosothiol (GSNO) as a nitric oxide (NO) donor loaded into calcium carbonate-based mineralized nanoparticles (GSNO-MNPs) to regulate cell signaling pathways for the osteogenic differentiation of mouse embryonic stem cells (ESCs). GSNO-MNPs were prepared by an anionic block copolymer template-mediated calcium carbonate (CaCO3) mineralization process in the presence of GSNO. GSNO-MNPs were spherical and had a narrow size distribution. GSNO was stably loaded within the MNPs without denaturation. TEM analysis also demonstrated the localization of GSNO-MNPs within membrane-bound structures in the cell, indicating the successful introduction of GSNO-MNPs into the cytosol of ESCs. Intracellular levels of NO and cGMP were significantly increased upon treatment with GSNO-MNPs, compared with the control group. When cells were exposed to GSNO-MNPs, the effects of nanoparticles on cell viability were not statistically significant. GSNO-MNPs treatment increased ALP activity assay and intracellular calcium levels. Real-time RT-PCR also revealed highly increased expression levels of the osteogenic target genes ALP, osteocalcin (OCN), and osterix (OSX) in GSNO-MNP-treated ESCs. The protein levels of OSX and Runt-related transcription factor 2 (RUNX2) showed similar patterns of expression based on real-time RT-PCR. These results indicate that GSNO-MNPs influenced the osteogenic differentiation of ESCs. Transcriptome profiling identified several significantly enriched and involved biological networks, such as RAP1, RAS, PI3K-AKT, and MAPK signaling pathways. These findings suggest that GSNO-MNPs can modulate osteogenic differentiation in ESCs via complex molecular pathways.
Collapse
Affiliation(s)
- Seong Yeong An
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hong Jae Lee
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sang Cheon Lee
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Jung Sun Heo
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
13
|
Suyasa IK, Wiradewi Lestari AA. Low expression of vascular endothelial growth factor and high serum level of cyclic guanine monophosphate as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rat. Chin J Traumatol 2020; 23:107-112. [PMID: 31980236 PMCID: PMC7156883 DOI: 10.1016/j.cjtee.2019.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Severe damage to the femoral head in patients with osteonecrosis has a high impact on morbidity. Despite early diagnosis, the treatment outcome is still unsatisfactory. This study aimed to explore the expression of vascular endothelial growth factor (VEGF) and cyclic guanine monophosphate (cGMP) serum level as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rats. METHODS This was an experimental study using randomized post-test only control group design, with samples using 10-14 weeks Wistar male rats. Rats were then divided into 6 groups: 3 groups without intervention, and 3 groups with intervention using 40% alcohol given perorally. Each one group from intervention and control group was euthanized by the end of the week for 3 consecutive weeks. Proximal femurs were examined under microscope for osteonecrosis, immunohistochemically for VEGF, and blood serum for cGMP levels. RESULTS VEGF expression in the femoral head of alcohol-exposed Wistar rats was lower than those not exposed to alcohol (p < 0.005). Blood serum cGMP levels of alcohol-exposed Wistar rats were higher than those not exposed to alcohol (p < 0.005). The number of necrotic osteocytes in the femoral head of Wistar rats exposed to alcohol was greater than those not exposed to alcohol (p < 0.005). There are significant differences between VEGF, cGMP levels, and number of necrotic osteocytes in the control group and treatment at 1st, 2nd, and 3rd week (p < 0.005). CONCLUSIONS Based on the result of this study, VEGF and cGMP may be considered as diagnostic biomarkers for alcohol-induced femoral head osteonecrosis.
Collapse
Affiliation(s)
- I Ketut Suyasa
- Department of Orthopedic and Traumatology, Faculty of Medicine, Udayana University, Bali, Indonesia
| | - Anak Agung Wiradewi Lestari
- Department of Clinical Pathology, Faculty of Medicine, Udayana University, Bali, Indonesia,Corresponding author.
| |
Collapse
|
14
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
15
|
Marzin P, Cormier-Daire V. New perspectives on the treatment of skeletal dysplasia. Ther Adv Endocrinol Metab 2020; 11:2042018820904016. [PMID: 32166011 PMCID: PMC7054735 DOI: 10.1177/2042018820904016] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
The last few decades have been marked by the identification of numerous genes implicated in genetic disorders, helping in the elucidation of the underlying pathophysiology of these conditions. This has allowed new therapeutic approaches to emerge such as cellular therapy, gene therapy, or pharmacological therapy for various conditions. Skeletal dysplasias are good models to illustrate these scientific advances. Indeed, several therapeutic strategies are currently being investigated in osteogenesis imperfecta; there are ongoing clinical trials based on pharmacological approaches, targeting signaling pathways in achondroplasia and fibrodysplasia ossificans progressiva or the endoplasmic reticulum stress in metaphyseal dysplasia type Schmid or pseudoachondroplasia. Moreover, the treatment of hypophosphatasia or Morquio A disease illustrates the efficacy of enzyme drug replacement. To provide a highly specialized multidisciplinary approach, these treatments are managed by reference centers. The emergence of treatments in skeletal dysplasia provides new perspectives on the prognosis of these severe conditions and may change prenatal counseling in these diseases over the coming years.
Collapse
Affiliation(s)
- Pauline Marzin
- Clinical Genetics, INSERM UMR 1163, Paris
Descartes-Sorbonne Paris Cité University, IMAGINE Institute, Necker Enfants
Malades Hospital, Paris, France
| | - Valérie Cormier-Daire
- Clinical Genetics, INSERM UMR 1163, Paris
Descartes-Sorbonne Paris Cité University, IMAGINE Institute, Necker Enfants
Malades Hospital, 149 rue de sevres, Paris, 75015, France
| |
Collapse
|
16
|
Hao ML, Wang GY, Zuo XQ, Qu CJ, Yao BC, Wang DL. Gut microbiota: an overlooked factor that plays a significant role in osteoporosis. J Int Med Res 2019; 47:4095-4103. [PMID: 31436117 PMCID: PMC6753565 DOI: 10.1177/0300060519860027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gut microbes are known as the body’s second gene pool. Symbiotic intestinal
bacteria play a major role in maintaining balance in humans. Bad eating habits,
antibiotic abuse, diseases, and a poor living environment have a negative effect
on intestinal flora. Abnormal intestinal microbes are prone to cause a variety
of diseases, affecting life expectancy and long-term quality of life, especially
in older people. Several recent studies have found a close association between
intestinal microorganisms and osteoporosis. The potential mechanism of
intestinal flora affecting bone formation or destruction by mediating nitric
oxide, the immune and endocrine systems, and other factors is briefly described
in this review. All of these factors may be responsible for the intestinal flora
that causes osteoporosis. Studying the relationship between intestinal flora and
bone health not only provides new ideas for studying the role of intestinal
microorganism in osteoporosis, but also provides a new therapeutic direction for
clinically refractory osteoporosis. Study of the relationship between intestinal
microbiota and osteoporosis is important for maintaining bone health and
minimizing osteoporosis.
Collapse
Affiliation(s)
- Meng-Lei Hao
- Department of Geriatric Medicine, Affiliated Hospital of Qinghai University, Xining, Qinghai Province, P.R. China
| | - Guang-Yao Wang
- Juxian Hospital of Traditional Chinese Medicine, Juxian, Shandong Provence, P.R. China
| | - Xiao-Qin Zuo
- Department of Geriatric Medicine, Affiliated Hospital of Qinghai University, Xining, Qinghai Province, P.R. China
| | - Chan-Juan Qu
- Department of Radiology, Peking Union Medical Hospital, Beijing, P.R. China
| | - Bo-Chen Yao
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, P. R. China
| | - Dong-Lai Wang
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Provence, P.R. China
| |
Collapse
|
17
|
Yue S, Whalen P, Jee YH. Genetic regulation of linear growth. Ann Pediatr Endocrinol Metab 2019; 24:2-14. [PMID: 30943674 PMCID: PMC6449614 DOI: 10.6065/apem.2019.24.1.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022] Open
Abstract
Linear growth occurs at the growth plate. Therefore, genetic defects that interfere with the normal function of the growth plate can cause linear growth disorders. Many genetic causes of growth disorders have already been identified in humans. However, recent genome-wide approaches have broadened our knowledge of the mechanisms of linear growth, not only providing novel monogenic causes of growth disorders but also revealing single nucleotide polymorphisms in genes that affect height in the general population. The genes identified as causative of linear growth disorders are heterogeneous, playing a role in various growth-regulating mechanisms including those involving the extracellular matrix, intracellular signaling, paracrine signaling, endocrine signaling, and epigenetic regulation. Understanding the underlying genetic defects in linear growth is important for clinicians and researchers in order to provide proper diagnoses, management, and genetic counseling, as well as to develop better treatment approaches for children with growth disorders.
Collapse
Affiliation(s)
- Shanna Yue
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Philip Whalen
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Youn Hee Jee
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA,Address for correspondence: Youn Hee Jee, MD Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA Tel: +1-301-435-5834 Fax: +1-301-402-0574 E-mail:
| |
Collapse
|
18
|
Nordberg RC, Wang H, Wu Q, Loboa EG. Corin is a key regulator of endochondral ossification and bone development via modulation of vascular endothelial growth factor A expression. J Tissue Eng Regen Med 2018; 12:2277-2286. [PMID: 30352487 DOI: 10.1002/term.2760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 08/21/2018] [Accepted: 10/18/2018] [Indexed: 01/06/2023]
Abstract
Corin has been studied extensively within the vascular system and is known to regulate blood pressure. We have shown that corin is one of the most highly upregulated genes during osteogenic differentiation of human adipose-derived stem cells (hASCs). This study tested the hypothesis that, through modulation of angiogenic signalling pathways, corin is a critical regulator of osteogenic differentiation and endochondral ossification. In vitro, corin expression in hASC was suppressed via siRNA knockdown and vascular endothelial growth factor A (VEGF-A) expression was quantified via reverse transcription polymerase chain reaction. In vivo, a murine corin knockout model (female, 10 weeks) was used to determine the effect of corin deficiency on long bone development. Wild-type and corin knockout long bones were compared via haematoxylin and eosin staining to assess tissue characteristics and cellular organization, three-point bending to assess mechanical characteristics, and immunohistochemistry to visualize VEGF-A expression patterns. Corin knockdown significantly (p < 0.05) increased VEGF-A mRNA expression during osteogenic differentiation. In vivo, corin knockout reduced tibial growth plate thickness (p < 0.01) and severely diminished the hypertrophic region. Corin knockout femurs had significantly increased stiffness (p < 0.01) and maximum loads (p < 0.01) but reduced postyield deflections (p < 0.01). In corin knockout mice, VEGF-A expression was increased near the growth plate but was reduced throughout the tibial shaft and distal head of the tibiae. This is the first study to show that corin is a key regulator of bone development by modulation of VEGF-A expression. Further elucidation of this mechanism will aid in the development of optimized bone tissue engineering and regenerative medicine therapies.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina
| | - Hao Wang
- Molecular Cardiology, Cleveland Clinic, Ohio
| | - Qingyu Wu
- Molecular Cardiology, Cleveland Clinic, Ohio
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina.,College of Engineering, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Wang L, Jia H, Tower RJ, Levine MA, Qin L. Analysis of short-term treatment with the phosphodiesterase type 5 inhibitor tadalafil on long bone development in young rats. Am J Physiol Endocrinol Metab 2018; 315:E446-E453. [PMID: 29920215 PMCID: PMC6230700 DOI: 10.1152/ajpendo.00130.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclic GMP (cGMP) is an important intracellular regulator of endochondral bone growth and skeletal remodeling. Tadalafil, an inhibitor of the phosphodiesterase (PDE) type 5 (PDE5) that specifically hydrolyzes cGMP, is increasingly used to treat children with pulmonary arterial hypertension (PAH), but the effect of tadalafil on bone growth and strength has not been previously investigated. In this study, we first analyzed the expression of transcripts encoding PDEs in primary cultures of chondrocytes from newborn rat epiphyses. We detected robust expression of PDE5 as the major phosphodiesterase hydrolyzing cGMP. Time-course experiments showed that C-type natriuretic peptide increased intracellular levels of cGMP in primary chondrocytes with a peak at 2 min, and in the presence of tadalafil the peak level of intracellular cGMP was 37% greater ( P < 0.01) and the decline was significantly attenuated. Next, we treated 1-mo-old Sprague Dawley rats with vehicle or tadalafil for 3 wk. Although 10 mg·kg-1·day-1 tadalafil led to a significant 52% ( P < 0.01) increase in tissue levels of cGMP and a 9% reduction ( P < 0.01) in bodyweight gain, it did not alter long bone length, cortical or trabecular bone properties, and histological features. In conclusion, our results indicate that PDE5 is highly expressed in growth plate chondrocytes, and short-term tadalafil treatment of growing rats at doses comparable to those used in children with PAH has neither obvious beneficial effect on long bone growth nor any observable adverse effect on growth plate structure and trabecular and cortical bone structure.
Collapse
Affiliation(s)
- Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Department of Orthopaedics, Shandong University Qilu Hospital, Shandong University , Jinan , China
| | - Haoruo Jia
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Department of Orthopaedics, The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, China
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Michael A Levine
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Division of Endocrinology and Diabetes and the Center for Bone Health, The Children's Hospital of Philadelphia , Philadelphia, Pennsylvania
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Chuchana P, Mausset-Bonnefont AL, Mathieu M, Espinoza F, Teigell M, Toupet K, Ripoll C, Djouad F, Noel D, Jorgensen C, Brondello JM. Secreted α-Klotho maintains cartilage tissue homeostasis by repressing NOS2 and ZIP8-MMP13 catabolic axis. Aging (Albany NY) 2018; 10:1442-1453. [PMID: 29920476 PMCID: PMC6046234 DOI: 10.18632/aging.101481] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/13/2018] [Indexed: 04/24/2023]
Abstract
Progressive loss of tissue homeostasis is a hallmark of numerous age-related pathologies, including osteoarthritis (OA). Accumulation of senescent chondrocytes in joints contributes to the age-dependent cartilage loss of functions through the production of hypertrophy-associated catabolic matrix-remodeling enzymes and pro-inflammatory cytokines. Here, we evaluated the effects of the secreted variant of the anti-aging hormone α-Klotho on cartilage homeostasis during both cartilage formation and OA development. First, we found that α-Klotho expression was detected during mouse limb development, and transiently expressed during in vitro chondrogenic differentiation of bone marrow-derived mesenchymal stem cells. Genome-wide gene array analysis of chondrocytes from OA patients revealed that incubation with recombinant secreted α-Klotho repressed expression of the NOS2 and ZIP8/MMP13 catabolic remodeling axis. Accordingly, α-Klotho expression was reduced in chronically IL1β-treated chondrocytes and in cartilage of an OA mouse model. Finally, in vivo intra-articular secreted α-Kotho gene transfer delays cartilage degradation in the OA mouse model. Altogether, our results reveal a new tissue homeostatic function for this anti-aging hormone in protecting against OA onset and progression.
Collapse
Affiliation(s)
- Paul Chuchana
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | - Marc Mathieu
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | | | - Karine Toupet
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | - Farida Djouad
- IRMB, INSERM, Montpellier University, Montpellier, France
| | - Danièle Noel
- IRMB, INSERM, Montpellier University, Montpellier, France
| | - Christian Jorgensen
- IRMB, INSERM, Montpellier University, Montpellier, France
- CHU Montpellier, Montpellier, France
| | | |
Collapse
|
21
|
Kalyanaraman H, Schall N, Pilz RB. Nitric oxide and cyclic GMP functions in bone. Nitric Oxide 2018; 76:62-70. [PMID: 29550520 PMCID: PMC9990405 DOI: 10.1016/j.niox.2018.03.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 01/24/2023]
Abstract
Nitric oxide plays a central role in the regulation of skeletal homeostasis. In cells of the osteoblastic lineage, NO is generated in response to mechanical stimulation and estrogen exposure. Via activation of soluble guanylyl cyclase (sGC) and cGMP-dependent protein kinases (PKGs), NO enhances proliferation, differentiation, and survival of bone-forming cells in the osteoblastic lineage. NO also regulates the differentiation and activity of bone-resorbing osteoclasts; here the effects are largely inhibitory and partly cGMP-independent. We review the skeletal phenotypes of mice deficient in NO synthases and PKGs, and the effects of NO and cGMP on bone formation and resorption. We examine the roles of NO and cGMP in bone adaptation to mechanical stimulation. Finally, we discuss preclinical and clinical data showing that NO donors and NO-independent sGC activators may protect against estrogen deficiency-induced bone loss. sGC represents an attractive target for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652, USA
| | - Nadine Schall
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652, USA
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0652, USA.
| |
Collapse
|
22
|
Yang S, Guo L, Su Y, Wen J, Du J, Li X, Liu Y, Feng J, Xie Y, Bai Y, Wang H, Liu Y. Nitric oxide balances osteoblast and adipocyte lineage differentiation via the JNK/MAPK signaling pathway in periodontal ligament stem cells. Stem Cell Res Ther 2018; 9:118. [PMID: 29716662 PMCID: PMC5930947 DOI: 10.1186/s13287-018-0869-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/28/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Critical tissues that undergo regeneration in periodontal tissue are of mesenchymal origin; thus, investigating the regulatory mechanisms underlying the fate of periodontal ligament stem cells could be beneficial for application in periodontal tissue regeneration. Nitric oxide (NO) regulates many biological processes in developing embryos and adult stem cells. The present study was designed to investigate the effects of NO on the function of human periodontal ligament stem cells (PDLSCs) as well as to elucidate the underlying molecular mechanisms. Methods Immunofluorescent staining and flow cytometry were used for stem cell identification. Western blot, reverse transcription polymerase chain reaction (RT-PCR), immunofluorescent staining, and flow cytometry were used to examine the expression of NO-synthesizing enzymes. The proliferative capacity of PDLSCs was determined by EdU assays. The osteogenic potential of PDLSCs was tested using alkaline phosphatase (ALP) staining, Alizarin Red staining, and calcium concentration detection. Oil Red O staining was used to analyze the adipogenic ability. Western blot, RT-PCR, and staining were used to examine the signaling pathway. Results Human PDLSCs expressed both inducible NO synthase (iNOS) and endothelial NO synthase (eNOS) and produced NO. Blocking the generation of NO with the NOS inhibitor l-NG-monomethyl arginine (l-NMMA) had no influence on PDLSC proliferation and apoptosis but significantly attenuated the osteogenic differentiation capacity and stimulated the adipogenic differentiation capacity of PDLSCs. Increasing the physiological level of NO with NO donor sodium nitroprusside (SNP) significantly promoted the osteogenic differentiation capacity but reduced the adipogenic differentiation capacity of PDLSCs. NO balances the osteoblast and adipocyte lineage differentiation in periodontal ligament stem cells via the c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) signaling pathway. Conclusions NO is essential for maintaining the balance between osteoblasts and adipocytes in PDLSCs via the JNK/MAPK signaling pathway. Graphical Abstract NO balances osteoblast and adipocyte lineage differentiation via JNK/MAPK signaling pathway![]() Electronic supplementary material The online version of this article (10.1186/s13287-018-0869-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shan Yang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Lijia Guo
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jing Wen
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yitong Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Jie Feng
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yongmei Xie
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yuxing Bai
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China.
| |
Collapse
|
23
|
Zhou H, Zhu J, Liu M, Wu Q, Dong N. Role of the protease corin in chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells. J Tissue Eng Regen Med 2017; 12:973-982. [PMID: 28714548 DOI: 10.1002/term.2514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/30/2017] [Accepted: 07/11/2017] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have the potency to differentiate into chondrocytes, osteocytes and adipocytes. Corin is a cardiac protease that activates the natriuretic peptides, thereby regulating blood volume and pressure. In addition to the heart, corin gene upregulation was reported in bone marrow- and adipose tissue-derived MSCs that underwent osteogenic differentiation. To date, the biological significance of corin expression in MSC differentiation remains unknown. In this study we isolated and cultured human bone marrow-derived MSCs that were capable of undergoing chondrogenic, osteogenic and adipogenic lineage differentiation. By reverse transcription polymerase chain reaction (RT-PCR) and immunostaining, we found that corin expression was upregulated when these MSCs underwent chondrogenic, osteogenic and adipogenic differentiation. The upregulation of corin expression was most significant in the cells undergoing chondrogenic lineage differentiation. Silencing corin gene expression by small hairpin RNA in the MSCs inhibited chondrogenic, but not osteogenic and adipogenic, differentiation. These results suggest a novel function of corin in MSC differentiation and chondrocyte development.
Collapse
Affiliation(s)
- Haibin Zhou
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinsong Zhu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
- Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Molecular therapeutic strategies for FGFR3 gene-related skeletal dysplasia. J Mol Med (Berl) 2017; 95:1303-1313. [PMID: 29063142 DOI: 10.1007/s00109-017-1602-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 12/24/2022]
Abstract
The FGFR3 gene encodes fibroblast growth factor receptor 3 protein, a negative regulator of chondrogenesis. Gain-of-function mutations result in constitutively activated FGFR3, leading to aberrant signal transduction, and accounting for inhibition of chondrocyte proliferation and differentiation. Generally, these pathogenic mutations maintain FGFR3 in an active state and cause diverse phenotypes in patients with skeletal dysplasia. For decades, studies have revealed the molecular mechanisms of constitutively activated FGFR3 and relevant therapeutic strategies. By modulating the FGFR3-induced signalling pathway with methods such as blocking binding between ligands and receptors, blocking tyrosine kinase activities, or antagonising the FGFR3 downstream signalling pathway, these strategies offer the possibility to ameliorate FGFR3 gene-related skeletal dysplasia phenotypes. In this review, we describe the mechanisms of potential therapeutic targets and underlying regulators and then systematically review molecular therapeutic strategies for FGFR3 gene-related skeletal dysplasia based on current knowledge.
Collapse
|
25
|
Wu LH, Zhang Q, Zhang S, Meng LY, Wang YC, Sheng CJ. Effects of gene knockdown of CNP on ventricular remodeling after myocardial ischemia-reperfusion injury through NPRB/Cgmp signaling pathway in rats. J Cell Biochem 2017; 119:1804-1818. [PMID: 28796407 DOI: 10.1002/jcb.26341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/08/2017] [Indexed: 01/27/2023]
Abstract
This study aimed to explore effects of CNP on ventricular remodeling following myocardial ischemia-reperfusion (I/R) injury through the NPRB/cGMP signaling pathway. Rat cardiomyocytes were assigned into: control, I/R, I/R + CNP, and I/R + 8-Br-cGMP groups. ELISA, qRT-PCR, and Western blotting were used to detect cGMP content and expression, respectively. After model establishment of I/R rats, normal control, CNP-/- control, I/R, and CNP-/- groups were set. Indexes of heart were detected using echocardiography and hemodynamics. ELISA was used to measure serum CNP, cGMP, LDH, cTn I, CK-MB, TNF-α, and IL-6 levels. Myocardial infarct was identified by TTC staining, and apoptosis conditions by TUNEL staining. QRT-PCR and Western blotting were adopted to detect expressions of CNP, NPRB, cGMP, and apoptosis-related genes. Compared with control group, cGMP contents and expression in the I/R, I/R + CNP and I/R + 8-Br-cGMP groups were decreased. Levels of LVEDV, LVESV, LVDS, LVDD, IVSD, LVM, LVEDP, and LVSP were higher in the I/R, CNP-/- control, and CNP-/- groups than normal control group while LVEF, SV, CO, and ±dp/dtmax were lower. Compared with the normal control group, LDH, cTn I, CK-MB, TNF-α, and IL-6 were higher in the I/R, CNP-/- control and CNP-/- groups; pathological changes and myocardial infarction were observed in the I/R, CNP-/- control, and CNP-/- groups; expressions of apoptosis-related genes in those groups were higher; while CNP, NPRB, cGMP, and Bcl-2 expressions were decreased. We came to the conclusion that gene knockdown of CNP blocks the NPRB/cGMP signaling pathway, thereby aggravating myocardial I/R injury and causing ventricular remodeling in rats.
Collapse
Affiliation(s)
- Lian-He Wu
- Department of Cardiac Surgery, Jining No.1 People's Hospital, Jining, China
| | - Qi Zhang
- Department of Cardiology, Dongying City People's Hospital, Dongying, China
| | - Shen Zhang
- Department of Cardiac Surgery, Jining No.1 People's Hospital, Jining, China
| | - Lu-Yu Meng
- Department of Cardiac Surgery, Jining No.1 People's Hospital, Jining, China
| | - Yan-Chi Wang
- Department of Cardiac Surgery, Jining No.1 People's Hospital, Jining, China
| | - Cun-Jian Sheng
- Department of Cardiac Surgery, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
26
|
Hoshino M, Kaneko K, Miyamoto Y, Yoshimura K, Suzuki D, Akaike T, Sawa T, Ida T, Fujii S, Ihara H, Tanaka J, Tsukuura R, Chikazu D, Mishima K, Baba K, Kamijo R. 8-Nitro-cGMP promotes bone growth through expansion of growth plate cartilage. Free Radic Biol Med 2017; 110:63-71. [PMID: 28559051 DOI: 10.1016/j.freeradbiomed.2017.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/06/2017] [Accepted: 05/25/2017] [Indexed: 01/29/2023]
Abstract
In endochondral ossification, growth of bones occurs at their growth plate cartilage. While it is known that nitric oxide (NO) synthases are required for proliferation of chondrocytes in growth plate cartilage and growth of bones, the precise mechanism by which NO facilitates these process has not been clarified yet. C-type natriuretic peptide (CNP) also positively regulate elongation of bones through expansion of the growth plate cartilage. Both NO and CNP are known to use cGMP as the second messenger. Recently, 8-nitro-cGMP was identified as a signaling molecule produced in the presence of NO in various types of cells. Here, we found that 8-nitro-cGMP is produced in proliferating chondrocytes in the growth plates, which was enhanced by CNP, in bones cultured ex vivo. In addition, 8-nitro-cGMP promoted bone growth with expansion of the proliferating zone as well as increase in the number of proliferating cells in the growth plates. 8-Nitro-cGMP also promoted the proliferation of chondrocytes in vitro. On the other hand, 8-bromo-cGMP enhanced the growth of bones with expansion of hypertrophic zone of the growth plates without affecting either the width of proliferating zone or proliferation of chondrocytes. These results indicate that 8-nitro-cGMP formed in growth plate cartilage accelerates chondrocyte proliferation and bone growth as a downstream molecule of NO.
Collapse
Affiliation(s)
- Marie Hoshino
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan; Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Kotaro Kaneko
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, Japan
| | - Yoichi Miyamoto
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan.
| | - Kentaro Yoshimura
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| | - Dai Suzuki
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoaki Ida
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigemoto Fujii
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
| | - Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| | - Risa Tsukuura
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, Japan
| | - Daichi Chikazu
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| | - Kazuyoshi Baba
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| |
Collapse
|
27
|
de Andrés MC, Takahashi A, Oreffo ROC. Demethylation of an NF-κB enhancer element orchestrates iNOS induction in osteoarthritis and is associated with altered chondrocyte cell cycle. Osteoarthritis Cartilage 2016; 24:1951-1960. [PMID: 27307355 DOI: 10.1016/j.joca.2016.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the methylation profile of the nuclear factor (NF)-κB enhancer region at -5.8 kb of inducible nitric oxide synthase (iNOS) and the subsequent role in the induction of osteoarthritis (OA) via cell cycle regulation. METHODS Percentage methylation was determined by pyrosequencing, gene expression by qRT-PCR and cell proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Transient transfections were induced to determine the effect of the NF-κB enhancer region on cell proliferation and the influence of DNA methylation. RESULTS In vitro de-methylation with 5-aza-dC showed decreased levels of DNA methylation at CpG sites localised at -5.8 kb, which correlated with higher levels of iNOS expression. In vitro methylation of the NF-κB enhancer region at -5.8 kb increased the percentage of cells at G0/G1 cell cycle phase. Loss of methylation within this region correlated with, enhanced proliferation and increased number of cells at G2/M phase. OA chondrocytes demonstrated up-regulation of the G0/G1 cell cycle progression markers Cyclin D1 and CDK6 in contrast to control cells. We demonstrate the loss of methylation that occurs at specific CpG sites localised at the -5.8 kb NF-κB enhancer region of the iNOS gene in OA chondrocytes permits the binding of this transcription factor activating the expression of iNOS. This results in subsequent altered cell cycle regulation, altered proliferative phenotype and transmission of the pathogenic phenotype to daughter cells. CONCLUSIONS This study indicates that inhibition of cell cycle progression by iNOS enhancer hypermethylation is capable of reducing pro-inflammatory responses via down-regulation of NF-κB with important therapeutic implications in OA.
Collapse
Affiliation(s)
- M C de Andrés
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK
| | - A Takahashi
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK; Department of Orthopaedic Surgery, Tohoku University Hospital, Sendai, Japan
| | - R O C Oreffo
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK.
| |
Collapse
|
28
|
Wit JM, Oostdijk W, Losekoot M, van Duyvenvoorde HA, Ruivenkamp CAL, Kant SG. MECHANISMS IN ENDOCRINOLOGY: Novel genetic causes of short stature. Eur J Endocrinol 2016; 174:R145-73. [PMID: 26578640 DOI: 10.1530/eje-15-0937] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/17/2022]
Abstract
The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure. In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate. We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFκB signalling, STAT3 and IGF2 have recently been discovered. Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations cause a syndromic form of short stature with elevated T3/T4 ratio. Disorders of signalling of various paracrine factors (FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in ∼3% of short children, and also rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal abnormalities, CNVs, and imprinting disorders.
Collapse
Affiliation(s)
- Jan M Wit
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Wilma Oostdijk
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Monique Losekoot
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Hermine A van Duyvenvoorde
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Claudia A L Ruivenkamp
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Sarina G Kant
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
29
|
Baron J, Sävendahl L, De Luca F, Dauber A, Phillip M, Wit JM, Nilsson O. Short and tall stature: a new paradigm emerges. Nat Rev Endocrinol 2015; 11:735-46. [PMID: 26437621 PMCID: PMC5002943 DOI: 10.1038/nrendo.2015.165] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In the past, the growth hormone (GH)-insulin-like growth factor 1 (IGF-1) axis was often considered to be the main system that regulated childhood growth and, therefore, determined short stature and tall stature. However, findings have now revealed that the GH-IGF-1 axis is just one of many regulatory systems that control chondrogenesis in the growth plate, which is the biological process that drives height gain. Consequently, normal growth in children depends not only on GH and IGF-1 but also on multiple hormones, paracrine factors, extracellular matrix molecules and intracellular proteins that regulate the activity of growth plate chondrocytes. Mutations in the genes that encode many of these local proteins cause short stature or tall stature. Similarly, genome-wide association studies have revealed that the normal variation in height seems to be largely due to genes outside the GH-IGF-1 axis that affect growth at the growth plate through a wide variety of mechanisms. These findings point to a new conceptual framework for understanding short and tall stature that is centred not on two particular hormones but rather on the growth plate, which is the structure responsible for height gain.
Collapse
Affiliation(s)
- Jeffrey Baron
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Lars Sävendahl
- Division of Pediatric Endocrinology, Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Francesco De Luca
- St. Christopher’s Hospital for Children, Section of Endocrinology and Diabetes; Drexel University College of Medicine, Department of Pediatrics, Philadelphia, PA, U.S.A
| | - Andrew Dauber
- Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, Division of Endocrinology, Cincinnati, Ohio, USA
| | - Moshe Phillip
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Children’s Diabetes, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Jan M. Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ola Nilsson
- Division of Pediatric Endocrinology, Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
- Center for Molecular Medicine, Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| |
Collapse
|
30
|
Wang W, Song MH, Miura K, Fujiwara M, Nawa N, Ohata Y, Kitaoka T, Kubota T, Namba N, Jin DK, Kim OH, Ozono K, Cho TJ. Acromesomelic dysplasia, type maroteaux caused by novel loss-of-function mutations of the NPR2 gene: Three case reports. Am J Med Genet A 2015; 170A:426-434. [PMID: 26567084 DOI: 10.1002/ajmg.a.37463] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/26/2015] [Indexed: 11/08/2022]
Abstract
The C-type natriuretic peptide (CNP)-natriuretic peptide receptor 2 (NPR2) signaling pathway plays an important role in chondrocyte development. Homozygous loss-of-function mutations of the NPR2 gene cause acromesomelic dysplasia, type Maroteaux (AMDM). The aim of this study was to identify and characterize NPR2 loss-of-function mutations in patients with AMDM. The NPR2 gene was sequenced in three Korean patients with AMDM and functional analysis of the mutated proteins was performed in vitro. Five novel NPR2 mutations were found in the three patients: two compound heterozygous mutations [c.1231T>C (Tyr411His) and c.2761C>T (Arg921X) in Patient 1 and c.1663A>T (Lys555X) and c.1711-1G>C (M571VfsX12) in Patient 3] and a homozygous mutation [c.2762G>A (Arg921Gln) in Patient 2]. Serum NT-proCNP concentration was significantly increased in each patient compared to control subjects. Cells transfected with the expression vector of each mutant except those found in Patient 3 showed a negligible or a markedly low cGMP response after treatment with CNP. HA-tagged wild-type (wt) and HA-mutant NPR2 were expressed at comparable levels: there were two bands of ∼130 and ∼120 kDa in wt and Arg921Gln, a single ∼120 kDa band in Tyr411His, and a single ∼110 kDa in the nonsense mutant. With respect to subcellular localization, Arg921Gln as well as wt-NPR2 reached the cell surface, whereas Tyr411His and Arg921X mutants did not. The Tyr411His and Arg921X NPR2 proteins were co-localized with an endoplasmic reticulum (ER) marker and failed to traffic from the ER to the Golgi apparatus. These results are consistent with deglycosylation experiments. Tyr411His and Arg921X NPR2 are complete loss-of-function mutations, whereas Arg921Gln behaves as a receptor for CNP with limited function.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mi Hyun Song
- Department of Orthopaedic Surgery, Jeju National University Hospital, Jeju, Republic of Korea
| | - Kohji Miura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Nobutoshi Nawa
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuhisa Ohata
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Taichi Kitaoka
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Noriyuki Namba
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Dong Kyu Jin
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ok Hwa Kim
- Department of Radiology, Woorisoa Children's Hospital, Seoul, Republic of Korea
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tae-Joon Cho
- Division of Pediatric Orthopaedics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| |
Collapse
|
31
|
Kaluđerović MR, Mojić M, Schreckenbach JP, Maksimović-Ivanić D, Graf HL, Mijatović S. A Key Role of Autophagy in Osteoblast Differentiation on Titanium-Based Dental Implants. Cells Tissues Organs 2015; 200:265-77. [PMID: 26316150 DOI: 10.1159/000434625] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2015] [Indexed: 11/19/2022] Open
Abstract
Autophagy plays an important role in embryogenesis, for the maintenance of tissue homeostasis and the elimination of damaged subcellular structures. Furthermore, autophagy could be a mode of physiological cell death and also be implicated in cell differentiation. Thus, we hypothesized that autophagy may have an impact on the differentiation of osteoblast cells influenced by various titanium-based surfaces. Interactions between smooth, commercially available pure titanium (Ti cp), rough Ticer, acid-etched Ti cp (SS) and M1-M3 (comprised of the monoclinic phase of sodium-titanium oxides and rutile; M2 contains amorphous calcium phosphates) and human osteoblast cells were investigated. Immunofluorescent staining was used for detecting autophagy, cell cluster formation and collagen type I (Col-1) expression. Flow cytometry was employed to identify autophagy, the production of endogenous nitric oxide (NO) and the size and granularity of the cells. Rough surfaces caused osteoblast differentiation via the autophagic-dependent PI3/Akt signalling pathway. These surfaces induced the formation of discrete populations of large, granular cells, i.e. mature osteoblasts. In addition, M1-M3 provoked the development of a third population of small, granular cells, responsible for cell cluster formation, which are important for the formation of bone noduli and mineralisation. The same surfaces induced faster osteoblast maturation and enhanced NO production, a hallmark of the already mentioned processes. Neither the mature osteoblasts nor the small cells appeared after the inhibition of autophagy. Inhibition of autophagy also prevented cell cluster formation. We demonstrate that autophagy plays an essential role in the osteoblast differentiation on titanium-based surfaces with rough topography.
Collapse
|
32
|
Peake NJ, Bader DL, Vessillier S, Ramachandran M, Salter DM, Hobbs AJ, Chowdhury TT. C-type natriuretic peptide signalling drives homeostatic effects in human chondrocytes. Biochem Biophys Res Commun 2015; 465:784-9. [PMID: 26307537 DOI: 10.1016/j.bbrc.2015.08.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
Abstract
Signals induced by mechanical loading and C-type natriuretic peptide (CNP) represent chondroprotective routes that may potentially prevent osteoarthritis (OA). We examined whether CNP will reduce hyaluronan production and export via members of the multidrug resistance protein (MRP) and diminish pro-inflammatory effects in human chondrocytes. The presence of interleukin-1β (IL-1β) increased HA production and export via MRP5 that was reduced with CNP and/or loading. Treatment with IL-1β conditioned medium increased production of catabolic mediators and the response was reduced with the hyaluronan inhibitor, Pep-1. The induction of pro-inflammatory cytokines by the conditioned medium was reduced by CNP and/or Pep-1, αCD44 or αTLR4 in a cytokine-dependent manner, suggesting that the CNP pathway is protective and should be exploited further.
Collapse
Affiliation(s)
- N J Peake
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - D L Bader
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - S Vessillier
- National Institute for Biological Standards and Control, Biotherapeutics Group, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - M Ramachandran
- Department of Orthopaedics and Trauma, The Royal London Hospital and Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Whitechapel Road, London E1 1BB, UK
| | - D M Salter
- Centre for Genomics and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crew Road, Edinburgh EH4 2XU, UK
| | - A J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, QMUL, Charterhouse Square, London EC1M 6BQ, UK
| | - T T Chowdhury
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| |
Collapse
|
33
|
Sun MMG, Beier F. Chondrocyte hypertrophy in skeletal development, growth, and disease. ACTA ACUST UNITED AC 2015; 102:74-82. [PMID: 24677724 DOI: 10.1002/bdrc.21062] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 12/31/2022]
Abstract
Most of our bones form through the process of endochondral ossification, which is tightly regulated by the activity of the cartilage growth plate. Chondrocyte maturation through the various stages of growth plate physiology ultimately results in hypertrophy. Chondrocyte hypertrophy is an essential contributor to longitudinal bone growth, but recent data suggest that these cells also play fundamental roles in signaling to other skeletal cells, thus coordinating endochondral ossification. On the other hand, ectopic hypertrophy of articular chondrocytes has been implicated in the pathogenesis of osteoarthritis. Thus, a better understanding of the processes that control chondrocyte hypertrophy in the growth plate as well as in articular cartilage is required for improved management of both skeletal growth disorders and osteoarthritis. This review summarizes recent findings on the regulation of hypertrophic chondrocyte differentiation, the cellular mechanisms involved in hypertrophy, and the role of chondrocyte hypertrophy in skeletal physiology and pathophysiology.
Collapse
Affiliation(s)
- Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Western University, and Children's Health Research Institute, London, Ontario, Canada
| | | |
Collapse
|
34
|
Irfanullah, Umair M, Khan S, Ahmad W. Homozygous Sequence Variants in theNPR2Gene Underlying Acromesomelic Dysplasia Maroteaux Type (AMDM) in Consanguineous Families. Ann Hum Genet 2015; 79:238-44. [DOI: 10.1111/ahg.12116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 03/23/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Irfanullah
- Department of Biochemistry, Faculty of Biological Sciences; Quaid-i-Azam University Islamabad; Pakistan
| | - Muhammad Umair
- Department of Biochemistry, Faculty of Biological Sciences; Quaid-i-Azam University Islamabad; Pakistan
| | - Saadullah Khan
- Department of Biotechnology & Genetic Engineering; Kohat University of Science & Technology; Kohat Khyber Pakhtunkhwa Pakistan
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences; Quaid-i-Azam University Islamabad; Pakistan
| |
Collapse
|
35
|
Cheung CSF, Zhu Z, Lui JCK, Dimitrov D, Baron J. Human monoclonal antibody fragments targeting matrilin-3 in growth plate cartilage. Pharm Res 2015; 32:2439-49. [PMID: 25690340 DOI: 10.1007/s11095-015-1636-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 01/21/2015] [Indexed: 11/26/2022]
Abstract
PURPOSE Many genetic disorders, including chondrodysplasias, and acquired disorders impair growth plate function, resulting in short and sometimes malformed bones. There are multiple endocrine and paracrine factors that promote chondrogenesis at the growth plate, which could potentially be used to treat these disorders. Targeting these growth factors specifically to the growth plate might augment the therapeutic skeletal effect while diminishing undesirable effects on non-target tissues. METHODS Using yeast display technology, we selected single-chain variable antibody fragments that bound to human and mouse matrilin-3, an extracellular matrix protein specifically expressed in cartilage tissue. The ability of the selected antibody fragments to bind matrilin-3 and to bind cartilage tissue in vitro and in vivo was assessed by ELISA and immunohistochemistry. RESULTS We identified antibody fragments that bound matrilin-3 with high affinity and also bound with high tissue specificity to cartilage homogenates and to cartilage structures in mouse embryo sections. When injected intravenously in mice, the antibody fragments specifically homed to cartilage. CONCLUSIONS Yeast display successfully selected antibody fragments that are able to target cartilage tissue in vivo. Coupling these antibodies to chondrogenic endocrine and paracrine signaling molecules has the potential to open up new pharmacological approaches to treat childhood skeletal growth disorders.
Collapse
Affiliation(s)
- Crystal Sao-Fong Cheung
- Section on Growth and Development, National Institute of Child Health and Development, National Institutes of Health, Bldg. 10 CRC, Rm. 1-3330, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | | | | | | | | |
Collapse
|
36
|
Shapiro F, Barone L, Johnson A, Flynn E. The cn/cn dwarf mouse. Histomorphometric, ultrastructural, and radiographic study in mutants corresponding to human acromesomelic dysplasia Maroteaux type (AMDM). BMC Musculoskelet Disord 2014; 15:347. [PMID: 25319082 PMCID: PMC4219045 DOI: 10.1186/1471-2474-15-347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 10/03/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The cn/cn dwarf mouse is caused by a loss-of-function mutation in the natriuretic peptide receptor 2 (NPR-2) gene which helps positively regulate endochondral longitudinal bone growth. The gene mutation corresponds to that in the human skeletal dysplasia Acromesomelic Dysplasia Maroteaux type (AMDM). This study assesses histomorphometric, ultrastructural and radiographic correlates of the growth abnormality. METHODS Ten litters of cn/cn and cn/+littermates at ages ranging from 2.5 to 6.5 weeks were studied by skeletal radiographs, histomorphometry and physeal ultrastructure. Skeletal radiographs were done on 2 cn/cn and 2 cn/+littermates at 5 weeks of age. Humeral, femoral, and tibial lengths were measured from 34 intact bones (17 cn/cn, 17 cn/+) at 2.5 to 6.5 weeks. Growth plate histomorphometry in 50 bones (26 cn/cn and 24 cn/+) determined the hypertrophic zone/entire physeal cartilage ratios in 204 sections (87 cn/+, 117 cn/cn) at 3 time periods (2.5-3, 4-4.5, and 6-6.5 weeks). Electron microscopy assessed 6 cn/cn and 6 cn/+age and site-matched physeal cartilage. RESULTS Cn/cn mice were two thirds the size of the cn/+. Cn/cn bones were normal in shape or only minimally deformed except for the radius with mid-diaphyseal bowing. Length ratios of cn/cn humeri, femurs, and tibias were a mean of 0.65 (± 0.03, n = 34, 17 ratios) compared to cn/+bones. The main physeal abnormality was a markedly shortened hypertrophic zone with the ratio of hypertrophic zone to entire physis 0.17 (± 0.063) in the cn/cn and 0.30 (± 0.052) in the cn/+mice. Ratio assessments were similar comparing humeral, femoral, and tibial growth plates as were ratios from each of the 3 time periods. Ultrastructural assessments from the resting zone to the lower hypertrophic zone-metaphyseal junction showed no specific individual cell abnormalities in cn/cn compared to cn/+physes. CONCLUSIONS The disorder causes a shortened physeal hypertrophic zone but normal ultrastructure of cn/cn chondrocytes points to abnormality primarily affecting the hypertrophic zone rather than a structural cell or matrix synthesis problem.
Collapse
Affiliation(s)
- Frederic Shapiro
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratory, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
37
|
Abstract
Corin is a serine protease originally isolated from the heart. Functional studies show that corin is the long-sought enzyme responsible for activating cardiac natriuretic peptides. In mice, lack of corin prevents natriuretic peptide processing, causing salt-sensitive hypertension. In humans, corin variants and mutations that reduce corin activity have been identified in patients with hypertension and heart failure. Decreased plasma levels of corin antigen and activity have been reported in patients with heart failure and coronary artery disease. Low levels of urinary corin also have been found in patients with chronic kidney disease. Most recent studies show that corin also acts in the uterus to promote spiral artery remodeling and prevent pregnancy-induced hypertension. Here, we review the role of corin in natriuretic peptide processing and cardiovascular diseases such as hypertension, heart disease, pre-eclampsia, and chronic kidney disease.
Collapse
|
38
|
Miura K, Kim OH, Lee HR, Namba N, Michigami T, Yoo WJ, Choi IH, Ozono K, Cho TJ. Overgrowth syndrome associated with a gain-of-function mutation of the natriuretic peptide receptor 2 (NPR2) gene. Am J Med Genet A 2013; 164A:156-63. [DOI: 10.1002/ajmg.a.36218] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 08/08/2013] [Indexed: 01/13/2023]
Affiliation(s)
- Kohji Miura
- Department of Pediatrics; Osaka University Graduate School of Medicine; Osaka Japan
| | - Ok-Hwa Kim
- Department of Radiology; Ajou University Hospital; Suwon Republic of Korea
| | - Hey Ran Lee
- Division of Pediatric Orthopaedics; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Noriyuki Namba
- Department of Pediatrics; Osaka University Graduate School of Medicine; Osaka Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research; Osaka Medical Centre and Research Institute for Maternal and Child Health; Osaka Japan
| | - Won Joon Yoo
- Division of Pediatric Orthopaedics; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - In Ho Choi
- Division of Pediatric Orthopaedics; Seoul National University Children's Hospital; Seoul Republic of Korea
| | - Keiichi Ozono
- Department of Pediatrics; Osaka University Graduate School of Medicine; Osaka Japan
| | - Tae-Joon Cho
- Division of Pediatric Orthopaedics; Seoul National University Children's Hospital; Seoul Republic of Korea
| |
Collapse
|
39
|
Michigami T. Regulatory mechanisms for the development of growth plate cartilage. Cell Mol Life Sci 2013; 70:4213-21. [PMID: 23640571 PMCID: PMC11113666 DOI: 10.1007/s00018-013-1346-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 04/11/2013] [Accepted: 04/15/2013] [Indexed: 12/26/2022]
Abstract
In vertebrates, most of the skeleton is formed through endochondral ossification. Endochondral bone formation is a complex process involving the mesenchymal condensation of undifferentiated cells, the proliferation of chondrocytes and their differentiation into hypertrophic chondrocytes, and mineralization. This process is tightly regulated by various factors including transcription factors, soluble mediators, extracellular matrices, and cell-cell and cell-matrix interactions. Defects of these factors often lead to skeletal dysplasias and short stature. Moreover, there is growing evidence that epigenetic and microRNA-mediated mechanisms also play critical roles in chondrogenesis. This review provides an overview of our current understanding of the regulators for the development of growth plate cartilage and their molecular mechanisms of action. A knowledge of the regulatory mechanisms underlying the proliferation and differentiation of chondrocytes will provide insights into future therapeutic options for skeletal disorders.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Osaka Medical Center and Research Institute for Maternal and Child Health, 840 Murodo-cho, Izumi, Osaka, 594-1101, Japan,
| |
Collapse
|
40
|
Mourskaia AA, Amir E, Dong Z, Tiedemann K, Cory S, Omeroglu A, Bertos N, Ouellet V, Clemons M, Scheffer GL, Park M, Hallett M, Komarova SV, Siegel PM. ABCC5 supports osteoclast formation and promotes breast cancer metastasis to bone. Breast Cancer Res 2012; 14:R149. [PMID: 23174366 PMCID: PMC4053136 DOI: 10.1186/bcr3361] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Accepted: 11/12/2012] [Indexed: 12/12/2022] Open
Abstract
Introduction Bone is the most common site of breast cancer metastasis, and complications associated with bone metastases can lead to a significantly decreased patient quality of life. Thus, it is essential to gain a better understanding of the molecular mechanisms that underlie the emergence and growth of breast cancer skeletal metastases. Methods To search for novel molecular mediators that influence breast cancer bone metastasis, we generated gene-expression profiles from laser-capture microdissected trephine biopsies of both breast cancer bone metastases and independent primary breast tumors that metastasized to bone. Bioinformatics analysis identified genes that are differentially expressed in breast cancer bone metastases compared with primary, bone-metastatic breast tumors. Results ABCC5, an ATP-dependent transporter, was found to be overexpressed in breast cancer osseous metastases relative to primary breast tumors. In addition, ABCC5 was significantly upregulated in human and mouse breast cancer cell lines with high bone-metastatic potential. Stable knockdown of ABCC5 substantially reduced bone metastatic burden and osteolytic bone destruction in mice. The decrease in osteolysis was further associated with diminished osteoclast numbers in vivo. Finally, conditioned media from breast cancer cells with reduced ABCC5 expression failed to induce in vitro osteoclastogenesis to the same extent as conditioned media from breast cancer cells expressing ABCC5. Conclusions Our data suggest that ABCC5 functions as a mediator of breast cancer skeletal metastasis. ABCC5 expression in breast cancer cells is important for efficient osteoclast-mediated bone resorption. Hence, ABCC5 may be a potential therapeutic target for breast cancer bone metastasis.
Collapse
|
41
|
An overgrowth disorder associated with excessive production of cGMP due to a gain-of-function mutation of the natriuretic peptide receptor 2 gene. PLoS One 2012; 7:e42180. [PMID: 22870295 PMCID: PMC3411678 DOI: 10.1371/journal.pone.0042180] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/02/2012] [Indexed: 01/12/2023] Open
Abstract
We describe a three-generation family with tall stature, scoliosis and macrodactyly of the great toes and a heterozygous p.Val883Met mutation in Npr2, the gene that encodes the CNP receptor NPR2 (natriuretic peptide receptor 2). When expressed in HEK293A cells, the mutant Npr2 cDNA generated intracellular cGMP (cyclic guanosine monophosphate) in the absence of CNP ligand. In the presence of CNP, cGMP production was greater in cells that had been transfected with the mutant Npr2 cDNA compared to wild-type cDNA. Transgenic mice in which the mutant Npr2 was expressed in chondrocytes driven by the promoter and intronic enhancer of the Col11a2 gene exhibited an enhanced production of cGMP in cartilage, leading to a similar phenotype to that observed in the patients. In addition, blood cGMP concentrations were elevated in the patients. These results indicate that p.Val883Met is a constitutive active gain-of-function mutation and elevated levels of cGMP in growth plates lead to the elongation of long bones. Our findings reveal a critical role for NPR2 in skeletal growth in both humans and mice, and may provide a potential target for prevention and treatment of diseases caused by impaired production of cGMP.
Collapse
|
42
|
Yan Q, Feng Q, Beier F. Reduced chondrocyte proliferation, earlier cell cycle exit and increased apoptosis in neuronal nitric oxide synthase-deficient mice. Osteoarthritis Cartilage 2012; 20:144-51. [PMID: 22179029 DOI: 10.1016/j.joca.2011.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 11/21/2011] [Accepted: 11/24/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Nitric oxide (NO) has been implicated in the local regulation of bone metabolism. However, the contribution made by specific nitric oxide synthase (NOS) enzymes to skeletal development is unclear. The objective of this study was to examine the effects of inactivation of neuronal nitric oxide synthase (nNOS) on cartilage development in mice. DESIGN Mice carrying a null mutation in the nNOS gene were used to address our objectives. Histological staining, immunohistochemistry and in situ analyses were employed along with real-time reverse transcriptase - polymerase chain reaction (RT-PCR). RESULTS nNOS-null mice show transient growth retardation and shorter long bones. nNOS-deficient growth plates show a reduction in replicating cells. Reduced chondrocyte numbers may in part be due to slower cell cycle progression and premature cell cycle exit caused by decreased cyclin D1 and increased p57 expression in mutants. In addition, apoptosis was increased as shown by increased cleaved-caspase 3 staining in hypertrophic chondrocytes in mutants. Real-time PCR demonstrated that expression of early chondrocyte markers such as Sox genes was reduced in mutant mice, while expression of prehypertrophic markers such as RORα was increased. Histological sections also demonstrated thinner cortical bone, fewer trabeculae and reduced mineralization in mutant mice. CONCLUSIONS These data identify an important role of nNOS in chondrocyte proliferation and endochondral bone growth and demonstrate that nNOS coordinates cell cycle exit and chondrocyte differentiation in cartilage development.
Collapse
Affiliation(s)
- Q Yan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
43
|
Wang G, Yan Q, Woods A, Aubrey LA, Feng Q, Beier F. Inducible nitric oxide synthase-nitric oxide signaling mediates the mitogenic activity of Rac1 during endochondral bone growth. J Cell Sci 2011; 124:3405-13. [PMID: 21965529 DOI: 10.1242/jcs.076026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Coordinated proliferation and differentiation of growth plate chondrocytes controls endochondral bone growth and final height in humans, and disruption of this process results in diseases of the growing and adult skeleton, such as chondrodysplasias or osteoarthritis. We had shown recently that chondrocyte-specific deletion of the gene Rac1 in mice leads to severe dwarfism due to reduced chondrocyte proliferation, but the molecular pathways involved remained unclear. Here, we demonstrate that Rac1-deficient chondrocytes have severely reduced levels of inducible nitric oxide synthase (iNOS) protein and nitric oxide (NO) production. NO donors reversed the proliferative effects induced by Rac1 deficiency, whereas inhibition of NO production mimicked the effects of Rac1 loss of function. Examination of the growth plate of iNOS-deficient mice revealed reduced chondrocyte proliferation and expression of cyclin D1, resembling the phenotype of Rac1-deficient growth plates. Finally, we demonstrate that Rac1-NO signaling inhibits the expression of ATF3, a known suppressor of cyclin D1 expression in chondrocytes. In conclusion, our studies identify the iNOS-NO pathway as a novel mediator of mitogenic Rac1 signaling and indicate that it could be a target for growth disorder therapies.
Collapse
Affiliation(s)
- Guoyan Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Lee EJ, Shin SH, Hyun S, Chun J, Kang SS. Mutation of a putative S-nitrosylation site of TRPV4 protein facilitates the channel activates. Anim Cells Syst (Seoul) 2011; 15:95-106. [PMID: 21837266 PMCID: PMC3150788 DOI: 10.1080/19768354.2011.555183] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/28/2010] [Accepted: 11/01/2010] [Indexed: 12/20/2022] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) cation channel, a member of the TRP vanilloid subfamily, is expressed in a broad range of tissues. Nitric oxide (NO) as a gaseous signal mediator shows a variety of important biological effects. In many instances, NO has been shown to exhibit its activities via a protein S-nitrosylation mechanism in order to regulate its protein functions. With functional assays via site-directed mutagenesis, we demonstrate herein that NO induces the S-nitrosylation of TRPV4 Ca2+ channel on the Cys853 residue, and the S-nitrosylation of Cys853 reduced its channel sensitivity to 4-α phorbol 12,13-didecanoate and the interaction between TRPV4 and calmodulin. A patch clamp experiment and Ca2+ image analysis show that the S-nitrosylation of Cys853 modulates the TRPV4 channel as an inhibitor. Thus, our data suggest a novel regulatory mechanism of TRPV4 via NO-mediated S-nitrosylation on its Cys853 residue.
Collapse
Affiliation(s)
- Eun Jeoung Lee
- Department of Pre-medicine, Eulji University School of Medicine, Daejeon 301-832, Republic of Korea
| | | | | | | | | |
Collapse
|
45
|
Gillespie JR, Ulici V, Dupuis H, Higgs A, Dimattia A, Patel S, Woodgett JR, Beier F. Deletion of glycogen synthase kinase-3β in cartilage results in up-regulation of glycogen synthase kinase-3α protein expression. Endocrinology 2011; 152:1755-66. [PMID: 21325041 DOI: 10.1210/en.2010-1412] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The rate of endochondral bone growth determines final height in humans and is tightly controlled. Glycogen synthase kinase-3 (GSK-3) is a negative regulator of several signaling pathways that govern bone growth, such as insulin/IGF and Wnt/β-catenin. The two GSK-3 proteins, GSK-3α and GSK-3β, display both overlapping and distinct roles in different tissues. Here we show that pharmacological inhibition of GSK-3 signaling in a mouse tibia organ culture system results in enhanced bone growth, accompanied by increased proliferation of growth plate chondrocytes and faster turnover of hypertrophic cartilage to bone. GSK-3 inhibition rescues some, but not all, effects of phosphatidylinositide 3-kinase inhibition in this system, in agreement with the antagonistic role of these two kinases in response to signals such as IGF. However, cartilage-specific deletion of the Gsk3b gene in mice has minimal effects on skeletal growth or development. Molecular analyses demonstrated that compensatory up-regulation of GSK-3α protein levels in cartilage is the likely cause for this lack of effect. To our knowledge, this is the first tissue in which such a compensatory mechanism is described. Thus, our study provides important new insights into both skeletal development and the biology of GSK-3 proteins.
Collapse
Affiliation(s)
- J R Gillespie
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lenz A, Bennett M, Skelton WP, Vesely DL. Vessel dilator and C-type natriuretic peptide enhance the proliferation of human osteoblasts. Pediatr Res 2010; 68:405-8. [PMID: 20613683 DOI: 10.1203/pdr.0b013e3181ef7636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
C-natriuretic peptide (CNP) has been shown to regulate proliferation of mouse and rat osteoblasts. Genetic deletion of CNP results in dwarfism. Overexposure of CNP has been associated with arachnodactyly of hands and feet with a very long hallux bilaterally in a 14-y-old girl. CNP effects on bone growth involve inhibition of MEK 1 and ERK 1/2 kinases mediated via the intracellular messenger cGMP. Vessel dilator is another natriuretic peptide synthesized by the atrial natriuretic peptide gene whose biologic half-life is 12 times longer than CNP. Vessel dilator's biologic effects on proliferating cells are mediated via inhibiting MEK 1/2 and ERK 1/2 kinases via cGMP. Vessel dilator has never been studied on osteoblasts. CNP at 10 (nanomolar) nM (p = 0.02) and vessel dilator at 10 nM, 1 nM, 100 (picomolar) pM, and 10 pM (p ≤ 0.01) in dose-response studies enhanced human osteoblasts' proliferation. This first study of human osteoblasts would suggest that vessel dilator with a much longer biologic half-life and with osteoblast-stimulatory effects at lower concentrations than CNP may have therapeutic potential in human achondroplasia, short stature, and osteoporosis. Vessel dilator stimulates osteoblast proliferation whereas most current therapies of osteoporosis target osteoclasts.
Collapse
Affiliation(s)
- Anne Lenz
- Department of Internal Medicine, University of South Florida School of Medicine, James A. Haley Veterans Medical Center, Tampa, Florida 33612, USA
| | | | | | | |
Collapse
|
47
|
Baldridge D, Shchelochkov O, Kelley, B, Lee B. Signaling Pathways in Human Skeletal Dysplasias. Annu Rev Genomics Hum Genet 2010; 11:189-217. [DOI: 10.1146/annurev-genom-082908-150158] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dustin Baldridge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030; , , ,
| | - Oleg Shchelochkov
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030; , , ,
- Department of Pediatrics, Division of Genetics, University of Iowa, Iowa City, Iowa 52242
| | - Brian Kelley,
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030; , , ,
- Howard Hughes Medical Institute, Houston, Texas 77009
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030; , , ,
- Howard Hughes Medical Institute, Houston, Texas 77009
| |
Collapse
|
48
|
Beier F, Loeser RF. Biology and pathology of Rho GTPase, PI-3 kinase-Akt, and MAP kinase signaling pathways in chondrocytes. J Cell Biochem 2010; 110:573-80. [PMID: 20512918 DOI: 10.1002/jcb.22604] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chondrocytes provide the framework for the developing skeleton and regulate long-bone growth through the activity of the growth plate. Chondrocytes in the articular cartilage, found at the ends of bones in diarthroidial joints, are responsible for maintenance of the tissue through synthesis and degradation of the extracellular matrix. The processes of growth, differentiation, cell death and matrix remodeling are regulated by a network of cell signaling pathways in response to a variety of extracellular stimuli. These stimuli consist of soluble ligands, including growth factors and cytokines, extracellular matrix proteins, and mechanical factors that act in concert to regulate chondrocyte function through a variety of canonical and non-canonical signaling pathways. Key chondrocyte signaling pathways include, but are not limited to, the p38, JNK and ERK MAP kinases, the PI-3 kinase-Akt pathway, the Jak-STAT pathway, Rho GTPases and Wnt-beta-catenin and Smad pathways. Modulation of the activity of any of these pathways has been associated with various pathological states in cartilage. This review focuses on the Rho GTPases, the PI-3 kinase-Akt pathway, and some selected aspects of MAP kinase signaling. Most studies to date have examined these pathways in isolation but it is becoming clear that there is significant cross-talk among the pathways and that the overall effects on chondrocyte function depend on the balance in activity of multiple signaling proteins.
Collapse
Affiliation(s)
- Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
49
|
Paradise WA, Vesper BJ, Goel A, Waltonen JD, Altman KW, Haines GK, Radosevich JA. Nitric oxide: perspectives and emerging studies of a well known cytotoxin. Int J Mol Sci 2010; 11:2715-45. [PMID: 20717533 PMCID: PMC2920563 DOI: 10.3390/ijms11072715] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 06/17/2010] [Accepted: 07/13/2010] [Indexed: 12/19/2022] Open
Abstract
The free radical nitric oxide (NO•) is known to play a dual role in human physiology and pathophysiology. At low levels, NO• can protect cells; however, at higher levels, NO• is a known cytotoxin, having been implicated in tumor angiogenesis and progression. While the majority of research devoted to understanding the role of NO• in cancer has to date been tissue-specific, we herein review underlying commonalities of NO• which may well exist among tumors arising from a variety of different sites. We also discuss the role of NO• in human physiology and pathophysiology, including the very important relationship between NO• and the glutathione-transferases, a class of protective enzymes involved in cellular protection. The emerging role of NO• in three main areas of epigenetics—DNA methylation, microRNAs, and histone modifications—is then discussed. Finally, we describe the recent development of a model cell line system in which human tumor cell lines were adapted to high NO• (HNO) levels. We anticipate that these HNO cell lines will serve as a useful tool in the ongoing efforts to better understand the role of NO• in cancer.
Collapse
Affiliation(s)
- William A. Paradise
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
| | - Benjamin J. Vesper
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
| | - Ajay Goel
- Division of Gastroenterology, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, Dallas, TX 75246, USA; E-Mail:
| | - Joshua D. Waltonen
- Department of Otolaryngology, Wake Forest University, Winston-Salem, NC 27157, USA; E-Mail:
| | | | - G. Kenneth Haines
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; E-Mail:
| | - James A. Radosevich
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA; E-Mails: (W.A.P.); (B.J.V.)
- Department of Jesse Brown, Veterans Administration Medical Center, Chicago, IL 60612, USA
- Author to whom correspondence should be addressed; E-Mail:
| |
Collapse
|
50
|
Shaikh N, Russo L, Papaleo E, Giannoni P, De Gioia L, Nicotra F, Quarto R, Cipolla L. C-type natriuretic peptide: Structural studies, fragment synthesis, and preliminary biological evaluation in human osteosarcoma cell lines. Biopolymers 2010; 94:213-9. [DOI: 10.1002/bip.21336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|