1
|
Vasylyev DV, Liu CJ, Waxman SG. Sodium channels in non-excitable cells: powerful actions and therapeutic targets beyond Hodgkin and Huxley. Trends Cell Biol 2025; 35:381-398. [PMID: 39743470 DOI: 10.1016/j.tcb.2024.11.009] [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: 09/03/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Voltage-gated sodium channels (VGSCs) are best known for their role in the generation and propagation of action potentials in neurons, muscle cells, and cardiac myocytes, which have traditionally been labeled as 'excitable'. However, emerging evidence challenges this traditional perspective. It is now clear that VGSCs are also expressed in a broad spectrum of cells outside the neuromuscular realm, where they regulate diverse cellular functions. In this review, we summarize current knowledge on the expression, regulation, and function of VGSCs in non-neuromuscular cells, highlighting their contributions to physiological processes and pathological conditions. Dynamic expression patterns of VGSCs in different cell types, involvement of VGSCs in cellular functions, such as phagocytosis, motility, and cytokine release, and their potential as therapeutic targets for diseases that include inflammatory disorders, osteoarthritis (OA), and cancer, are discussed. This new understanding of VGSCs and their effects on cells outside the neuromuscular realm opens new avenues for research and therapeutic interventions.
Collapse
Affiliation(s)
- Dmytro V Vasylyev
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.
| |
Collapse
|
2
|
Dewdney B, Miranda PJ, Kuchibhotla M, Palanisamy R, Richworth C, Milligan CJ, Ng ZY, Ursich L, Petrou S, Fletcher EV, Daly RJ, Lim Kam Sian TCC, Valvi S, Endersby R, Johns TG. Ion channel modulator DPI-201-106 significantly enhances antitumor activity of DNA damage response inhibitors in glioblastoma. Neurooncol Adv 2024; 6:vdae187. [PMID: 39659830 PMCID: PMC11630809 DOI: 10.1093/noajnl/vdae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Background Glioblastoma, a lethal high-grade glioma, has not seen improvements in clinical outcomes in nearly 30 years. Ion channels are increasingly associated with tumorigenesis, and there are hundreds of brain-penetrant drugs that inhibit ion channels, representing an untapped therapeutic resource. The aim of this exploratory drug study was to screen an ion channel drug library against patient-derived glioblastoma cells to identify new treatments for brain cancer. Methods Seventy-two ion channel inhibitors were screened in patient-derived glioblastoma cells, and cell viability was determined using the ViaLight Assay. Cell cycle and apoptosis analysis were determined with flow cytometry using PI and Annexin V staining, respectively. Protein and phosphoprotein expression was determined using mass spectrometry and analyzed using gene set enrichment analysis. Kaplan-Meier survival analyses were performed using intracranial xenograft models of GBM6 and WK1 cells. Results The voltage-gated sodium channel modulator, DPI-201-106, was revealed to reduce glioblastoma cell viability in vitro by inducing cell cycle arrest and apoptosis. Phosphoproteomics indicated that DPI-201-106 may impact DNA damage response (DDR) pathways. Combination treatment of DPI-201-106 with the CHK1 inhibitor prexasertib or the PARP inhibitor niraparib demonstrated synergistic effects in multiple patient-derived glioblastoma cells both in vitro and in intracranial xenograft mouse models, extending survival of glioblastoma-bearing mice. Conclusions DPI-201-106 enhances the efficacy of DDR inhibitors to reduce glioblastoma growth. As these drugs have already been clinically tested in humans, repurposing DPI-201-106 in novel combinatorial approaches will allow for rapid translation into the clinic.
Collapse
Affiliation(s)
- Brittany Dewdney
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Panimaya Jeffreena Miranda
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Mani Kuchibhotla
- The Kids Research Institute, Perth, Western Australia, Australia
| | | | | | - Carol J Milligan
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zi Ying Ng
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Lauren Ursich
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Steve Petrou
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emily V Fletcher
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Terry C C Lim Kam Sian
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Santosh Valvi
- Perth Children’s Hospital, Perth, Western Australia, Australia
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Raelene Endersby
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| | - Terrance G Johns
- Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia
- The Kids Research Institute, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Wang H, Liu Z, Niu D, Li H, Han Y, Peng J, Qian Q. Carbamazepine regulates USP10 through miR-20a-5p to affect the deubiquitination of SKP2 and inhibit osteogenic differentiation. J Orthop Surg Res 2023; 18:820. [PMID: 37915040 PMCID: PMC10619296 DOI: 10.1186/s13018-023-04169-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/07/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Antiepileptic drugs (AEDs) harm bone health and are significantly associated with osteoporosis development. In this study, we aimed to explore the mechanisms involved in carbamazepine (CBZ) and microRNA (miR)-20a-5p/ubiquitin-specific peptidase 10 (USP10)/S-phase kinase-associated protein 2 (SKP2) axis in osteoporosis. METHODS Human bone marrow mesenchymal stem cells (BMSCs) were treated with different concentrations of CBZ. Knocking down or overexpressing miR-20a-5p, USP10, and SKP2 cell lines were constructed. The expressions of miR-20a-5p, USP10, SKP2, runt-related transcription factor 2 (Runx2), Alkaline phosphatase (ALP), Osterix (Osx), osteocalcin (OCN) and Collagen I were detected with western blot (WB) and reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR). Alizarin Red S (ARS) staining was performed to measure calcium deposition. Dual-luciferase assay and RNA immunoprecipitation (RIP) were applied to verify the binding relationship between miR-20a-5p and USP10. USP10 and SKP2 combination was verified by Co-Immunopurification (Co-IP). The stability of the SKP2 protein was verified by Cycloheximide chase assay. RESULTS CBZ could reduce cell activity. ALP activity and ARS staining were enhanced in the osteogenic induction (OM) group. The expressions of Runx2, ALP, Osx, OCN and Collagen I were increased. CBZ reduced miR-20a-5p expressions. Verification experiments showed miR-20a-5p could target USP10. USP10 increased SKP2 stability and promoted SKP2 expression. CBZ regulated miR-20a-5p/USP10/SPK2 and inhibited BMSCs osteogenic differentiation. CONCLUSIONS CBZ regulated USP10 through miR-20a-5p to affect the deubiquitination of SKP2 and inhibit osteogenic differentiation, which provided a new idea for osteoporosis treatment.
Collapse
Affiliation(s)
- Huan Wang
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Ziye Liu
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Dawei Niu
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
- Department of Orthopedics, No. 971 Hospital of the PLA Navy, Qingdao, 266071, People's Republic of China
| | - Haobo Li
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Jinhui Peng
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China.
| | - Qirong Qian
- Department of Orthopedics, Shanghai Changzheng Hospital, No.415, Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
4
|
Terrone G, Gragnaniello V, Esposito A, Del Puente A, Del Giudice E. Effects of antiepileptic therapy on bone mineral status evaluated by phalangeal quantitative ultrasound in pediatric patients with epilepsy and motor impairment. Minerva Pediatr (Torino) 2023; 75:476-481. [PMID: 31129950 DOI: 10.23736/s2724-5276.18.05235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
BACKGROUND In epileptic patients with motor disability, it's difficult to disentangle the effects of antiepileptic drugs (AEDs) on bone health from those provoked by impaired mobility. The aim of this study was to evaluate the effects of AEDs on bone mineral status by phalangeal quantitative ultrasound (QUS), a no-radiation and non-invasive method, in pediatric patients with motor impairment and epilepsy. METHODS We enrolled 56 patients (31 females, 25 males) with epilepsy and motor impairment and 24 children with only motor disability (13 females, 11 males). Patients were stratified by Gross Motor Function Classification System Scale (GMFCS) in 4 groups: group A1 with epilepsy and mild motor impairment (GMFCS levels I-II), group A2 with only mild motor impairment, group B1 with epilepsy and severe motor impairment (GMFCS levels III-V), group B2 with only severe motor impairment. The bone mineral status was evaluated by phalangeal QUS and amplitude-dependent speed of sound (AD-SoS) Z-score was calculated for each patient. RESULTS The four groups showed no significant differences in age, gender and 25-hydroxyvitamin D levels. The group B1 had a statistically lower amplitude-dependent speed of sound Z-score as compared to group A2 (P<0.05). The multivariate analysis of independent factors revealed a significant correlation between amplitude-dependent speed of sound Z-score and Gross Motor Function Classification System levels (P=0.004). The mean Z-score value decreased by 0.53, increasing the motor impairment. CONCLUSIONS The bone mineral status measured as AD-SoS strongly correlates with severity of motor disability evaluated by GMFCS as compared to antiepileptic therapy and 25-hydroxyvitamin D levels.
Collapse
Affiliation(s)
- Gaetano Terrone
- Section of Pediatrics, Department of Translational Medicine, University of Naples Federico II, Naples, Italy -
| | - Vincenza Gragnaniello
- Section of Pediatrics, Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Antonella Esposito
- Unit of Rheumatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Antonio Del Puente
- Unit of Rheumatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ennio Del Giudice
- Section of Pediatrics, Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Wu PM, Cho HY, Chiang CW, Chuang TH, Wu SN, Tu YF. Characterization in Inhibitory Effectiveness of Carbamazepine in Voltage-Gated Na + and Erg-Mediated K + Currents in a Mouse Neural Crest-Derived (Neuro-2a) Cell Line. Int J Mol Sci 2022; 23:7892. [PMID: 35887240 PMCID: PMC9321339 DOI: 10.3390/ijms23147892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 12/18/2022] Open
Abstract
Carbamazepine (CBZ, Tegretol®) is an anticonvulsant used in the treatment of epilepsy and neuropathic pain; however, several unwanted effects of this drug have been noticed. Therefore, the regulatory actions of CBZ on ionic currents in electrically excitable cells need to be reappraised, although its efficacy in suppressing voltage-gated Na+ current (INa) has been disclosed. This study was undertaken to explore the modifications produced by CBZ on ionic currents (e.g., INa and erg-mediated K+ current [IK(erg)]) measured from Neuro-2a (N2a) cells. In these cells, we found that this drug differentially suppressed the peak (transient, INa(T)) and sustained (late, INa(L)) components of INa in a concentration-dependent manner with effective IC50 of 56 and 18 μM, respectively. The overall current-voltage relationship of INa(T) with or without the addition of CBZ remained unchanged; however, the strength (i.e., ∆area) in the window component of INa (INa(W)) evoked by the short ascending ramp pulse (Vramp) was overly lessened in the CBZ presence. Tefluthrin (Tef), a synthetic pyrethroid, known to stimulate INa, augmented the strength of the voltage-dependent hysteresis (Hys(V)) of persistent INa (INa(P)) in response to the isosceles-triangular Vramp; moreover, further application of CBZ attenuated Tef-mediated accentuation of INa(P)'s Hys(V). With a two-step voltage protocol, the recovery of INa(T) inactivation seen in Neuro-2a cells became progressively slowed by adding CBZ; however, the cumulative inhibition of INa(T) evoked by pulse train stimulation was enhanced during exposure to this drug. Neuro-2a-cell exposure to CBZ (100 μM), the magnitude of erg-mediated K+ current measured throughout the entire voltage-clamp steps applied was mildly inhibited. The docking results regarding the interaction of CBZ and voltage-gate Na+ (NaV) channel predicted the ability of CBZ to bind to some amino-acid residues in NaV due to the existence of a hydrogen bond or hydrophobic contact. It is conceivable from the current investigations that the INa (INa(T), INa(L), INa(W), and INa(P)) residing in Neuro-2a cells are susceptible to being suppressed by CBZ, and that its block on INa(L) is larger than that on INa(T). Collectively, the magnitude and gating of NaV channels produced by the CBZ presence might have an impact on its anticonvulsant and analgesic effects occurring in vivo.
Collapse
Affiliation(s)
- Po-Ming Wu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Tzu-Hsien Chuang
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
6
|
Pagel CN, Kularathna PK, Sanaei R, Young ND, Hooper JD, Mackie EJ. Protease-activated receptor-2 dependent and independent responses of bone cells to prostate cancer cell secretory products. Prostate 2022; 82:723-739. [PMID: 35167724 DOI: 10.1002/pros.24316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/25/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Metastatic prostate cancer lesions in the skeleton are frequently characterized by excessive formation of bone. Prostate cancer cells secrete factors, including serine proteases, that are capable of influencing the behavior of surrounding cells. Some of these proteases activate protease-activated receptor-2 (PAR2 ), which is expressed by osteoblasts (bone-forming cells) and precursors of osteoclasts (bone-resorbing cells). The aim of the current study was to investigate a possible role for PAR2 in regulating the behavior of bone cells exposed to metastatic prostate cancer cells. METHODS The effect of medium conditioned by the PC3, DU145, and MDA-PCa-2b prostate cancer cell lines was investigated in assays of bone cell function using cells isolated from wildtype and PAR2 -null mice. Osteoclast differentiation was assessed by counting tartrate-resistant acid phosphatase-positive multinucleate cells in bone marrow cultured in osteoclastogenic medium. Osteoblasts were isolated from calvariae of neonatal mice, and BrdU incorporation was used to assess their proliferation. Assays of alkaline phosphatase activity and quantitative PCR analysis of osteoblastic gene expression were used to assess osteoblast differentiation. Responses of osteoblasts to medium conditioned by MDA-PCa-2b cells were analyzed by RNAseq. RESULTS Conditioned medium (CM) from all three cell lines inhibited osteoclast differentiation independently of PAR2 . Media from PC3 and DU145 cells had no effect on assays of osteoblast function. Medium conditioned by MDA-PCa-2b cells stimulated BrdU incorporation in both wildtype and PAR2 -null osteoblasts but increased alkaline phosphatase activity and Runx2 and Col1a1 expression in wildtype but not PAR2 -null cells. Functional enrichment analysis of RNAseq data identified enrichment of multiple gene ontology terms associated with lysosomal function in both wildtype and PAR2 -null cells in response to MDA-PCa-2b-CM. Analysis of individual genes identified osteogenesis-associated genes that were either upregulated by MDA-PCa-2b-CM selectively in wildtype cells or downregulated selectively in PAR2 -null cells. CONCLUSIONS Factors secreted by prostate cancer cells influence bone cell behavior through both PAR2 -dependent and -independent mechanisms. Both PAR2 -independent suppression of osteoclast differentiation and PAR2 -dependent stimulation of osteogenesis are likely to determine the nature of prostate cancer metastases in bone.
Collapse
Affiliation(s)
- Charles N Pagel
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Pamu K Kularathna
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Reza Sanaei
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - John D Hooper
- Mater Research Institute, Translational Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Eleanor J Mackie
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Wagener N, Di Fazio P, Böker KO, Matziolis G. Osteogenic Effect of Pregabalin in Human Primary Mesenchymal Stem Cells, Osteoblasts, and Osteosarcoma Cells. Life (Basel) 2022; 12:496. [PMID: 35454987 PMCID: PMC9032037 DOI: 10.3390/life12040496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/06/2022] [Accepted: 03/26/2022] [Indexed: 02/07/2023] Open
Abstract
Seventy million patients worldwide are suffering from epilepsy. The long-term use of antiepileptic drugs causes the alteration of the bone tissue and its metabolism, thus increasing the risk of fractures. Clinical and pre-clinical studies have highlighted conflicting data on the influence of the relatively new antiepileptic drug pregabalin (Lyrica®). The objective of the present study was therefore to investigate its cytotoxicity in primary human osteoblasts (hOB). HOB and human mesenchymal stem cells (hMSC) were isolated from patients. The human osteosarcoma cells MG63 were included as established cell line. Cells were incubated with pregabalin at concentrations ranging from 0 to 40 μg/mL. Time-dependent cell proliferation was measured by automatic cell counting, and metabolism was determined by XTT assay and osseous differentiation by alkaline phosphatase (ALP) activity. Histological examinations of calcium deposit were performed with ALP, Alizarin Red, and von Kossa staining. A concentration-dependent increase in the proliferation of hOB and hMSC was observed after treatment with pregabalin. All cells showed a significant increase in cell metabolism. The osteogenic differentiation, confirmed by the increase of calcium deposit, was promoted by the administration of pregabalin. This effect was already significant at the therapeutic plasma concentration of pregabalin (10 μg/mL). In contrast to the other antiepileptic drugs, pregabalin showed no osteocatabolic effects. Conflicting in-vivo data must therefore be attributed to systemic effects of pregabalin.
Collapse
Affiliation(s)
- Nele Wagener
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany;
| | - Pietro Di Fazio
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany;
| | - Kai Oliver Böker
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany;
| | - Georg Matziolis
- Orthopaedic Professorship of the University Hospital Jena, Orthopaedic Department Waldkliniken Eisenberg, 07607 Eisenberg, Germany;
| |
Collapse
|
8
|
Abstract
Genetic mutations have long been implicated in epilepsy, particularly in genes that encode ion channels and neurotransmitter receptors. Among some of those identified are voltage-gated sodium, potassium and calcium channels, and ligand-gated gamma-aminobutyric acid (GABA), neuronal nicotinic acetylcholine (CHRN), and glutamate receptors, making them key therapeutic targets. In this chapter we discuss the use of automated electrophysiological technologies to examine the impact of gene defects in two potassium channels associated with different epilepsy syndromes. The hKCNC1 gene encodes the voltage-gated potassium channel hKV3.1, and mutations in this gene cause progressive myoclonus epilepsy (PME) and ataxia due to a potassium channel mutation (MEAK). The hKCNT1 gene encodes the weakly voltage-dependent sodium-activated potassium channel hKCNT1, and mutations in this gene cause a wide spectrum of seizure disorders, including severe autosomal dominant sleep-related hypermotor epilepsy (ADSHE) and epilepsy of infancy with migrating focal seizures (EIMFS), both conditions associated with drug-resistance. Importantly, both of these potassium channels play vital roles in regulating neuronal excitability. Since its discovery in the late nineteen seventies, the patch-clamp technique has been regarded as the bench-mark technology for exploring ion channel characteristics. In more recent times, innovations in automated patch-clamp technologies, of which there are many, are enabling the study of ion channels with much greater productivity that manual systems are capable of. Here we describe aspects of Nanion NPC-16 Patchliner, examining the effects of temperature on stably and transiently transfected mammalian cells, the latter of which for most automated systems on the market is quite challenging. Remarkable breakthroughs in the development of other automated electrophysiological technologies, such as multielectrode arrays that support extracellular signal recordings, provide additional features to examine network activity in the area of ion channel research, particularly epilepsy. Both of these automated technologies enable the acquisition of consistent, robust, and reproducible data. Numerous systems have been developed with very similar capabilities, however, not all the systems on the market are adapted to work with primary cells, particularly neurons that can be problematic. This chapter also showcases methods that demonstrate the versatility of Nanion NPC-16 Patchliner and the Multi Channel Systems (MCS) multielectrode array (MEA) assay for acutely dissociated murine primary cortical neurons, enabling the study of potassium channel mutations implicated in severe refractory epilepsies.
Collapse
|
9
|
Park YA, Subasinghe AK, Ahmad BS, Gorelik A, Garland SM, Clifford V, Chiang C, Robinson H, Wark JD. Associations Between Serum Sodium Concentration and Bone Health Measures in Individuals Who Use Antiepileptic Drugs: A Pilot Study. J Clin Densitom 2020; 23:364-372. [PMID: 31036448 DOI: 10.1016/j.jocd.2019.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Yeung-Ae Park
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Australia.
| | - Asvini K Subasinghe
- Royal Women's Hospital, Department of Microbiology and Infectious Diseases, Parkville, Australia; Murdoch Children's Research Institute, Infection and Immunity Theme, Parkville, Australia
| | - Baemisla Shiek Ahmad
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - Alexandra Gorelik
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Australia; School of Psychology, Australian Catholic University, Fitzroy, Australia
| | - Suzanne M Garland
- Royal Women's Hospital, Department of Microbiology and Infectious Diseases, Parkville, Australia; Murdoch Children's Research Institute, Infection and Immunity Theme, Parkville, Australia; University of Melbourne, Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Australia
| | - Vanessa Clifford
- Royal Women's Hospital, Department of Microbiology and Infectious Diseases, Parkville, Australia; Murdoch Children's Research Institute, Infection and Immunity Theme, Parkville, Australia; Department of Pathology, Royal Melbourne Hospital, Parkville, Australia
| | - Cherie Chiang
- Department of Pathology, Royal Melbourne Hospital, Parkville, Australia
| | - Heather Robinson
- Bone and Mineral Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - John D Wark
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Australia; Bone and Mineral Medicine, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
10
|
Siniscalchi A, Murphy S, Cione E, Piro L, Sarro GD, Gallelli L. Antiepileptic Drugs and Bone Health: Current Concepts. PSYCHOPHARMACOLOGY BULLETIN 2020; 50:36-44. [PMID: 32508365 PMCID: PMC7255839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Chronic use of antiepileptic drugs (AEDs) can induce the development of adverse effects on bone metabolism. In epileptic patients treated with AED, the monitoring of biochemical markers of bone turnover, such as the measurement of serum 25 (OH) vitamin D, bone mineral density, before the beginning of the treatment and during the follow-up is not routinely required. In the future, monitoring of biochemical markers in epileptic patients treated with AED may help us for adequate prevention therapy.
Collapse
Affiliation(s)
- Antonio Siniscalchi
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Sean Murphy
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Erika Cione
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Leonardo Piro
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Giovambattista De Sarro
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Luca Gallelli
- Siniscalchi, Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy. Murphy, General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland. Cione, Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), Italy. Piro, Orthopedic Unit, Corigliano-Rossano Hospital, ASP Cosenza, Italy. De Sarro and Gallelli, Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| |
Collapse
|
11
|
Automated Planar Patch-Clamp Recording of P2X Receptors. Methods Mol Biol 2020; 2041:285-300. [PMID: 31646497 DOI: 10.1007/978-1-4939-9717-6_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
P2X receptors are a structurally and functionally distinctive family of ligand-gated ion channels that play important roles in mediating extracellular adenosine 5'-triphosphate (ATP) signaling in diverse physiological and pathophysiological processes. For several decades, the "manual" patch-clamp technique was regarded as the gold standard assay for investigating ion channel properties. More recently, breakthroughs in the development of automated patch-clamp technologies are enabling the study of ion channels, with much greater throughput capacities. These automated platforms, of which there are many, generate consistent, reliable, high-fidelity data. This chapter demonstrates the versatility of one of these technologies for ligand-gated ion channels, with a particular emphasis on protocols that address some of the issues of receptor desensitization that are commonly associated with P2X receptor-mediated currents.
Collapse
|
12
|
Coates BA, McKenzie JA, Buettmann EG, Liu X, Gontarz PM, Zhang B, Silva MJ. Transcriptional profiling of intramembranous and endochondral ossification after fracture in mice. Bone 2019; 127:577-591. [PMID: 31369916 PMCID: PMC6708791 DOI: 10.1016/j.bone.2019.07.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/27/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
Abstract
Bone fracture repair represents an important clinical challenge with nearly 1 million non-union fractures occurring annually in the U.S. Gene expression differs between non-union and healthy repair, suggesting there is a pattern of gene expression that is indicative of optimal repair. Despite this, the gene expression profile of fracture repair remains incompletely understood. In this work, we used RNA-seq of two well-established murine fracture models to describe gene expression of intramembranous and endochondral bone formation. We used top differentially expressed genes, enriched gene ontology terms and pathways, callus cellular phenotyping, and histology to describe and contrast these bone formation processes across time. Intramembranous repair, as modeled by ulnar stress fracture, and endochondral repair, as modeled by femur full fracture, exhibited vastly different transcriptional profiles throughout repair. Stress fracture healing had enriched differentially expressed genes associated with bone repair and osteoblasts, highlighting the strong osteogenic repair process of this model. Interestingly, the PI3K-Akt signaling pathway was one of only a few pathways uniquely enriched in stress fracture repair. Full fracture repair involved a higher level of inflammatory and immune cell related genes than did stress fracture repair. Full fracture repair also differed from stress fracture in a robust downregulation of ion channel genes following injury, the role of which in fracture repair is unclear. This study offers a broad description of gene expression in intramembranous and endochondral ossification across several time points throughout repair and suggests several potentially intriguing genes, pathways, and cells whose role in fracture repair requires further study.
Collapse
Affiliation(s)
- Brandon A Coates
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America.
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| | - Xiaochen Liu
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Paul M Gontarz
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Bo Zhang
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| |
Collapse
|
13
|
Garip Ustaoglu S, Evis Z, Ilbay G, Boskey AL, Severcan F. Side-Effects of Convulsive Seizures and Anti-Seizure Therapy on Bone in a Rat Model of Epilepsy. APPLIED SPECTROSCOPY 2018; 72:689-705. [PMID: 28905646 DOI: 10.1177/0003702817734617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The severe sole effects of seizures on the cortical part of bone were reported in our previous study. However, the side effects of anti-epileptic drug therapy on bones has not been differentiated from the effects of the convulsive seizures, yet. This study provides the first report on differentiation of the effects of seizures and carbamazepine (a widely used antiepileptic drug) therapy on bones; 50 mg/kg/day drug was given to genetically induced absence epileptic rats for five weeks. Distinct bone regions including cortical, trabecular, and growth plate in each of tibia, femur, and spine tissues were studied using Fourier transform infrared (FT-IR) imaging and Vickers microhardness test. Blood levels of vitamin D and bone turnover biomarkers were also measured. According to the FT-IR imaging results, both seizure and carbamazepine-treated groups, more dominantly the drug-treated group, had lower mineral content with altered collagen crosslinks and higher crystallinity, implying reduced bone strength. Lower microhardness values also supported lower mechanical strength in bones. The most affected bone tissue and region from seizures and treatment was found as the spine and cortical, respectively. While there was a reduction in vitamin D and calcium levels in both seizure and carbamazepin-treated groups, significantly elevated PTH and bone turnover biomarkers were only seen in the drug-treated group.
Collapse
Affiliation(s)
- Sebnem Garip Ustaoglu
- 1 Department of Biochemistry, 187458 Middle East Technical University , Ankara, Turkey
- 2 Department of Medical Biochemistry, 187458 Faculty of Medicine, Altinbas University, Istanbul, Turkey
| | - Zafer Evis
- 3 Department of Engineering Sciences, Middle East Technical University, Ankara, Turkey
| | - Gul Ilbay
- 4 Department of Physiology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Adele Ludin Boskey
- 5 25062 Mineralized Tissue Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Feride Severcan
- 6 Department of Biological Sciences, 187458 Middle East Technical University , Ankara, Turkey
- 7 Department of Biophysics, 187458 Faculty of Medicine, Altinbas University, Istanbul, Turkey
| |
Collapse
|
14
|
Durá-Travé T, Gallinas-Victoriano F, Malumbres-Chacón M, Moreno-Gónzalez P, Aguilera-Albesa S, Yoldi-Petri ME. Vitamin D deficiency in children with epilepsy taking valproate and levetiracetam as monotherapy. Epilepsy Res 2018; 139:80-84. [DOI: 10.1016/j.eplepsyres.2017.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/16/2017] [Accepted: 11/27/2017] [Indexed: 01/25/2023]
|
15
|
Abstract
PURPOSE OF REVIEW It is well-recognized that individuals with epilepsy have an increased risk of vertebral and nonvertebral fractures; this increased risk has been described to be secondary to an increased bone fragility and to an increased risk of falls. Osteoporosis is the most common bone disease which has been characterized by microarchitectural deterioration of trabecula and cortical bone mass with a decrease in bone mineral density and bone strength. Specific side effects of antiepileptic drugs (AEDs) on bone metabolism have been identified; recent research publications further characterized some of the specific side effects of AEDs on bone metabolism. It is the purpose of this review to describe recent advances on the knowledge of the effects of AEDs on bone metabolism and the cause of osteoporosis in the field of epilepsy. RECENT FINDINGS Recent literature demonstrates that the increased risk of fractures in the epileptic patient population is likely multifactorial and includes seizure activity, injuries from falls, decreased bone strength, adverse effects from AEDs. Reviewed publications suggest that the mechanism of adverse effects on bone metabolism may differ among different AEDs. The impact of vitamin D deficiency or its metabolism in the epileptic population has also been a concern of several reviewed publications. SUMMARY This is a review is of the recent epilepsy and osteoporosis literature published over the past 18 months, highlighting reports and studies concerning the cause, pathogenesis, and possible preventive measures and effects of AEDs on changes of bone metabolism, bone loss, and development of osteoporosis. In addition, we also reviewed articles focusing on issues of prevention and treatment of osteoporosis in individuals with epilepsy. We utilized the search engines of PubMed and Cochrane Reviews from January 2016 to June 2017.
Collapse
Affiliation(s)
- Philip M Dussault
- aVA Boston Healthcare System bVA Boston Health Care System and Boston University School of Medicine and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
16
|
Simm PJ, Seah S, Gorelik A, Gilbert L, Nuguid J, Werther GA, Mackay MT, Freeman JL, Petty SJ, Wark JD. Impaired bone and muscle development in young people treated with antiepileptic drugs. Epilepsia 2017; 58:1931-1938. [DOI: 10.1111/epi.13893] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Peter J. Simm
- Department of Endocrinology and Diabetes; Royal Children's Hospital; Melbourne Victoria Australia
- Murdoch Childrens Research Institute; Melbourne Victoria Australia
- Department of Paediatrics; University of Melbourne; Melbourne Victoria Australia
| | - Sebastian Seah
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
| | - Alex Gorelik
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
- Melbourne EpiCentre; Royal Melbourne Hospital; Parkville Victoria Australia
| | - Lauren Gilbert
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
| | - Jenning Nuguid
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
| | - George A. Werther
- Department of Endocrinology and Diabetes; Royal Children's Hospital; Melbourne Victoria Australia
- Murdoch Childrens Research Institute; Melbourne Victoria Australia
- Department of Paediatrics; University of Melbourne; Melbourne Victoria Australia
| | - Mark T. Mackay
- Murdoch Childrens Research Institute; Melbourne Victoria Australia
- Department of Paediatrics; University of Melbourne; Melbourne Victoria Australia
- Department of Neurology; Royal Children's Hospital; Melbourne Victoria Australia
| | - Jeremy L. Freeman
- Murdoch Childrens Research Institute; Melbourne Victoria Australia
- Department of Neurology; Royal Children's Hospital; Melbourne Victoria Australia
| | - Sandra J. Petty
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
- Melbourne Brain Centre at Royal Melbourne Hospital; Melbourne Victoria Australia
- Academic Centre; Ormond College; Parkville Victoria Australia
| | - John D. Wark
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Victoria Australia
- Bone & Mineral Medicine; Royal Melbourne Hospital; Melbourne Victoria Australia
| |
Collapse
|
17
|
Neuron subset-specific Pten deletion induces abnormal skeletal activity in mice. Exp Neurol 2017; 291:98-105. [PMID: 28163158 DOI: 10.1016/j.expneurol.2017.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/27/2017] [Accepted: 02/01/2017] [Indexed: 12/13/2022]
Abstract
Individuals with a history of epilepsy are at higher risk for bone fractures compared to the general population. Although clinical studies support an association between low bone mineral density (BMD) and anti-seizure medications, little is known on whether a history of seizures is linked to altered bone health. Therefore, in this study we tested the hypothesis that bone mass, morphology, and bone mineralization are altered by seizures in genetically epileptic animals and in animals subjected to an episode of status epilepticus. In this study, we used NS-Pten conditional knockout mice (a well-studied genetic model of epilepsy). We used microCT analysis to measure BMD, morphology, and mineralization in NS-Pten+/+ (wildtype) and NS-Pten-/- (knockout) mice at 4 and 8weeks, as well as adult Kv4.2+/+ and Kv4.2-/- mice. We measured BMD, bone morphology, and mineralization in adult NS-Pten+/+ mice that received status epilepticus through kainic acid (20mg/kg intraperitoneal). Further, we measured locomotion for NS-Pten+/+ and NS-Pten-/- mice at 4 and 6weeks. We found that NS-Pten-/- mice exhibited low BMD in the tibial metaphysis and midshaft compared to non-epileptic mice. Morphologically, NS-Pten-/- mice exhibited decreased trabecular volume fraction, and endocortical expansion in both the metaphyeal and diaphyseal compartments. In the midshaft, NS-Pten-/- mice exhibited reduced tissue mineral density, indicating impaired mineralization in addition to morphological deficits. NS-Pten-/- mice exhibited hyperactivity in open field testing, suggesting low bone mass in NS-Pten-/- mice was not attributable to hypoactivity. Differences in BMD were not observed following kainate-induced seizures or in the Kv4.2-/- model of seizure susceptibility. Our findings suggest that deletion of Pten in the brain results in impaired bone mass and mineralization, which may contribute to weaker bones and thereby a higher fracture risk.
Collapse
|