1
|
Zillinger LS, Liesegang A, Hustedt K, Schnepel N, Sauerwein H, Schmicke M, Schwennen C, Muscher-Banse AS. Influence of N- and/or P-restriction on bone metabolism in young goats. Br J Nutr 2024; 132:874-886. [PMID: 39402760 PMCID: PMC11576093 DOI: 10.1017/s0007114524002150] [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: 02/22/2024] [Revised: 07/31/2024] [Accepted: 08/30/2024] [Indexed: 11/20/2024]
Abstract
Ruminants can recycle nitrogen (N) and phosphorus (P), which are essential for vital body processes. Reduced N- and P-intake in ruminants is desirable for economic and ecologic reasons. Simultaneous modulation of mineral homoeostasis and bone metabolism occurs in young goats. This study aimed to investigate potential effects of dietary N- and/or P-restriction on molecular changes in bone metabolism. The twenty-eight young male goats were fed a control diet, an N-reduced diet, a P-reduced diet or a combined N- and P-reduced diet for 6-8 weeks. The N-restricted goats had lower plasma Ca concentration and higher plasma osteocalcin (OC) and CrossLaps concentrations. The P-restricted goats had reduced plasma inorganic phosphate (Pi) concentrations and increased plasma Ca concentrations. Due to the initiation of a signalling pathway that inhibits the fibroblast growth factor 23 (FGF23) expression, this was lower with P-restriction. Consequently, lower Pi concentrations were the main factor influencing the reduction in FGF23. The changes in mineral homoeostasis associated with P-restriction led to a reduction in OC, bone mineral content and mineral density. Simultaneously, bone resorption potentially increased with P-restriction as indicated by an increased receptor activator of NF-κB ligand/osteoprotegerin (OPG) ratio and an increase in OPG mRNA expression. Additionally, the increased mRNA expression of the calcitonin receptor during P-restriction points to a higher number of osteoclasts. This study demonstrates an impairment of bone remodelling processes in young goats by N- or P-restriction. With P-restriction, bone mineralisation rate was potentially reduced and bone quality impaired, while with N-restriction, bone remodelling increased.
Collapse
Affiliation(s)
- Luisa S Zillinger
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Annette Liesegang
- Institute of Animal Nutrition and Dietetics, Vetsuisse Faculty Zurich, University of Zurich, Zurich8057, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), Zurich, Switzerland
| | - Karin Hustedt
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn53115, Germany
| | - Marion Schmicke
- Clinic for Diseases of Cattle, University of Veterinary Medicine Hannover Foundation, Hannover30173, Germany
| | - Cornelia Schwennen
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover Foundation, Hannover30173, Germany
| | - Alexandra S Muscher-Banse
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| |
Collapse
|
2
|
Venkatesh VS, Nie T, Golub S, Stok KS, Hemmatian H, Desai R, Handelsman DJ, Zajac JD, Grossmann M, Davey RA. High circulating concentrations of estradiol are anabolic for bone mass and strength in an adult male to female transgender mouse model. Bone 2024; 186:117143. [PMID: 38866125 DOI: 10.1016/j.bone.2024.117143] [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: 03/25/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
The effects of gender affirming hormone therapy (GAHT) on bone microarchitecture and fracture risk in adult transgender women is unclear. To investigate the concept that skeletal integrity and strength in trans women may be improved by treatment with a higher dose of GAHT than commonly prescribed, we treated adult male mice with a sustained, high dose of estradiol. Adult male mice at 16 weeks of age were administered ~1.3 mg estradiol by silastic implant, implanted intraperitoneally, for 12 weeks. Controls included vehicle treated intact females and males. High-dose estradiol treatment in males stimulated the endocortical deposition of bone at the femoral mid-diaphysis, increasing cortical thickness and bone area. This led to higher stiffness, maximum force, and the work required to fracture the bone compared to male controls, while post-yield displacement was unaffected. Assessment of the material properties of the bone showed an increase in both elastic modulus and ultimate stress in the estradiol treated males. Treatment of male mice with high dose estradiol was also anabolic for trabecular bone, markedly increasing trabecular bone volume, number and thickness in the distal metaphysis which was accompanied by an increase in the histomorphometric markers of bone remodelling, mineralizing surface/bone surface, bone formation rate and osteoclast number. In conclusion, a high dose of estradiol is anabolic for cortical and trabecular bone in a male to female transgender mouse model, increasing both stiffness and strength. These findings suggest that increasing the current dose of GAHT administered to trans women, while considering other potential adverse effects, may be beneficial to preserving their bone microstructure and strength.
Collapse
Affiliation(s)
- Varun S Venkatesh
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia; Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia.
| | - Tian Nie
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| | - Suzanne Golub
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| | - Kathryn S Stok
- Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Haniyeh Hemmatian
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| | - Reena Desai
- ANZAC Research Institute, University of Sydney and Andrology, Concord Repatriation General Hospital, Concord, New South Wales 2137, Australia.
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney and Andrology, Concord Repatriation General Hospital, Concord, New South Wales 2137, Australia.
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| | - Mathis Grossmann
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia; Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia.
| | - Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| |
Collapse
|
3
|
Becker LL, Gebhardt JT, Tokach MD, Woodworth JC, Goodband RD, DeRouchey JM. A review of calcium and phosphorus requirement estimates for gestating and lactating sows. Transl Anim Sci 2024; 8:txae087. [PMID: 38863597 PMCID: PMC11165643 DOI: 10.1093/tas/txae087] [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: 03/22/2024] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
Calcium (Ca) and phosphorus (P) are minerals involved in biological functions and essential structural components of the skeleton. The body tightly regulates Ca and P to maintain homeostasis. Maternal needs for Ca and P increase during gestation and lactation to support conceptus growth and milk synthesis. Litter size and litter average daily gain (ADG) have a large effect on Ca and P requirements for sows because as they increase, the requirements increase due to a greater need from the sow. The objective of this review was to summarize published literature on Ca and P requirements in gestating and lactating sows derived from empirical data and factorial models. A total of nine empirical studies and seven factorial models were reviewed for determining the Ca and P requirements in gestation. For lactation, there were six empirical studies and seven factorial models reviewed. Empirical studies determined requirements based on the observed effect of Ca and P on bone mineralization, sow and litter performance, and milk characteristics. Factorial models generated equations to estimate Ca and P requirements using the main components of maintenance, fetal and placental growth, and maternal retention in gestation. The main components for factorial equations in lactation include maintenance and milk production. In gestation, the standardized total tract digestible phosphorus (STTD P) requirement estimates from empirical studies range from 5.4 to 9.5 g/d with total Ca ranging from 12.9 to 18.6 g/d to maximize bone measurements or performance criteria. According to the factorial models, the requirements increase throughout gestation to meet the needs of the growing fetuses and range from 7.6 to 10.6 g/d and 18.4 to 38.2 g/d of STTD P and total Ca, respectively, on day 114 of gestation for parity 1 sows. During lactation, STTD P requirement estimates from empirical studies ranged from 8.5 to 22.1 g/d and total Ca ranged from 21.2 to 50.4 g/d. For the lactation factorial models, STTD P requirements ranged from 14.2 to 25.1 g/d for STTD P and 28.4 to 55.6 g/d for total Ca for parity 1 sows with a litter size of 15 pigs. The large variation in requirement estimates makes it difficult to define Ca and P requirements; however, a minimum level of 6.0 and 22.1 g/d of STTD P during gestation and lactation, respectively, appears to be adequate to meet basal requirements. The limited data and high variation indicate a need for future research evaluating Ca and P requirements for gestating and lactating sows.
Collapse
Affiliation(s)
- Larissa L Becker
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS, USA
| | - Jordan T Gebhardt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Mike D Tokach
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS, USA
| | - Jason C Woodworth
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS, USA
| | - Robert D Goodband
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS, USA
| | - Joel M DeRouchey
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
4
|
Li Z, Wen X, Li N, Zhong C, Chen L, Zhang F, Zhang G, Lyu A, Liu J. The roles of hepatokine and osteokine in liver-bone crosstalk: Advance in basic and clinical aspects. Front Endocrinol (Lausanne) 2023; 14:1149233. [PMID: 37091847 PMCID: PMC10117885 DOI: 10.3389/fendo.2023.1149233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Both the liver and bone are important secretory organs in the endocrine system. By secreting organ factors (hepatokines), the liver regulates the activity of other organs. Similarly, bone-derived factors, osteokines, are created during bone metabolism and act in an endocrine manner. Generally, the dysregulation of hepatokines is frequently accompanied by changes in bone mass, and osteokines can also disrupt liver metabolism. The crosstalk between the liver and bone, particularly the function and mechanism of hepatokines and osteokines, has increasingly gained notoriety as a topic of interest in recent years. Here, based on preclinical and clinical evidence, we summarize the potential roles of hepatokines and osteokines in liver-bone interaction, discuss the current shortcomings and contradictions, and make recommendations for future research.
Collapse
Affiliation(s)
- Zhanghao Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
| | - Xiaoxin Wen
- Department of Anatomy, Jinzhou Medical University, Jinzhou, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
| | - Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
| | - Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- *Correspondence: Jin Liu, ; Aiping Lyu,
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- *Correspondence: Jin Liu, ; Aiping Lyu,
| |
Collapse
|
5
|
Wei FL, Gao QY, Zhu KL, Heng W, Du MR, Yang F, Gao HR, Li T, Qian JX, Zhou CP. Efficacy and safety of pharmacologic therapies for prevention of osteoporotic vertebral fractures in postmenopausal women. Heliyon 2023; 9:e11880. [PMID: 36852077 PMCID: PMC9958453 DOI: 10.1016/j.heliyon.2022.e11880] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/22/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND There are many pharmaceutical interventions available to prevent osteoporotic vertebral fractures in postmenopausal women, but the efficacy and safety of these drugs are unknown. This study aimed to investigate the efficacy and safety of drugs in the prevention of osteoporotic vertebral fractures. METHODS PubMed, Embase, and the Cochrane Library were comprehensively searched for randomized controlled trials (RCTs) published up to February 15, 2020, including postmenopausal women with osteoporosis. Network meta-analysis was conducted based on the Cochrane Handbook for Systematic Reviews of Interventions and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The relative risk (RR) and 95% confidence interval (CI) were used to report the results. This study was registered with PROSPERO, number CRD42020201167. Main Outcomes were incidences of new vertebral fracture and serious adverse events. RESULTS Fifty-five RCTs (n = 104 580) evaluating vertebral fractures of sixteen kinds of pharmacologic therapies were included in the network meta-analysis. Abaloparatide (RR, 0.21; [95% CI, 0.09 to 0.51]), alendronate (RR, 0.55; [95% CI, 0.38 to 0.81]), calcitonin (RR, 0.44; [95% CI, 0.25 to 0.78]), denosumab (RR, 0.33; [95% CI, 0.14 to 0.61]), parathyroid hormone (PTH) (RR, 0.32; [95% CI, 0.10 to 0.97]), risedronate (RR, 0.65; [95% CI, 0.42 to 1.00]), romosozumab (RR, 0.31; [95% CI, 0.16 to 0.61]), strontium ranelate (RR, 0.62; [95% CI, 0.42 to 0.93]), teriparatide (RR, 0.27; [95% CI, 0.17 to 0.43]), and zoledronate (RR, 0.41; [95% CI, 0.93]) were associated with lower vertebral fracture risk compared to placebo. PTH was associated with more adverse event rates. For any two drug treatments, the RR of serious adverse events was not statistically significant. Hormone replacement therapy (HRT) and calcitonin may be slower to work because they have only been shown to reduce the risk of vertebral fractures in long-term (>18 months) follow-up. CONCLUSIONS A variety of drugs are safe and effective in preventing osteoporotic vertebral fractures. HRT and calcitonin only reduced the risk of vertebral fractures during a follow-up of 21-72 months.
Collapse
Affiliation(s)
- Fei-Long Wei
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Quan-You Gao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Kai-Long Zhu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Wei Heng
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Ming-Rui Du
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Fan Yang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Hao-Ran Gao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Ji-Xian Qian
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| | - Cheng-Pei Zhou
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038, Xi'an, China
| |
Collapse
|
6
|
Young K, Beggs MR, Grimbly C, Alexander RT. Regulation of 1 and 24 hydroxylation of vitamin D metabolites in the proximal tubule. Exp Biol Med (Maywood) 2022; 247:1103-1111. [PMID: 35482362 PMCID: PMC9335508 DOI: 10.1177/15353702221091982] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Calcium and phosphate are critical for numerous physiological processes. Consequently, the plasma concentration of these ions are tightly regulated. Calcitriol, the active form of vitamin D, is a positive modulator of mineralization as well as calcium and phosphate metabolism. The molecular and physiological effects of calcitriol are well documented. Calcitriol increases blood calcium and phosphate levels by increasing absorption from the intestine, and resorption of bone. Calcitriol synthesis is a multistep process. A precursor is first made via skin exposure to UV, it is then 25-hydroxylated in the liver to form 25-hydroxyitamin D. The next hydroxylation step occurs in the renal proximal tubule via the 1-αhydroxylase enzyme (encoded by CYP27B1) thereby generating 1,25-dihydroxyvitamin D, that is, calcitriol. At the same site, the 25-hydroxyvitamin D 24-hydroxlase enzyme encoded by CYP24A1 can hydroxylate 25-hydroxyvitamin D or calcitriol to deactivate the hormone. Plasma calcitriol levels are primarily determined by the regulated expression of CYP27B1 and CYP24A1. This occurs in response to parathyroid hormone (increases CYP27B1), calcitriol itself (decreases CYP27B1 and increases CYP24A1), calcitonin (increases or decreases CYP24A1 and increases CYP27B1), FGF23 (decreases CYP27B1 and increases CYP24A1) and potentially plasma calcium and phosphate levels themselves (mixed effects). Herein, we review the regulation of CYP27B1 and CYP24A1 transcription in response to the action of classic phophocalciotropic hormones and explore the possibility of direct regulation by plasma calcium.
Collapse
Affiliation(s)
- Kennedi Young
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada,Women and Children’s Health Institute, Edmonton, AB T6G 1C9, Canada
| | - Megan R Beggs
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada,Women and Children’s Health Institute, Edmonton, AB T6G 1C9, Canada
| | - Chelsey Grimbly
- Department of Paediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - R Todd Alexander
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada,Women and Children’s Health Institute, Edmonton, AB T6G 1C9, Canada,Department of Paediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada,R Todd Alexander.
| |
Collapse
|
7
|
L GB, JG H, I LT, M JM, P BÁ. The Sperm Olfactory Receptor OLFR601 is Dispensable for Mouse Fertilization. Front Cell Dev Biol 2022; 10:854115. [PMID: 35721474 PMCID: PMC9204177 DOI: 10.3389/fcell.2022.854115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
Fertilization involves the fusion of two gametes by means of yet unknown membrane binding and fusion events. Over the last years, many sperm proteins have been uncovered to play essential roles in sperm-egg fusion in mammals, but their precise role in fertilization remains unknown, being unclear how these proteins interact with each other or with other yet unknown sperm proteins. The aim of this study has been to identify possible sperm proteins interacting with TMEM95, a protein essential for fertilization located in the sperm membrane. A list of 41 sperm proteins that were pulled down with TMEM95 and identified by mass spectrometry did not include other sperm proteins known to play a role in fertilization, suggesting an independent role of TMEM95 in fertilization. Between these lists, OLFR601 is allocated to the acrosomal region and may mediate affinity for an odorant involved in fertilization. However, Olfr601 disruption did not impair the sperm fertilization ability, suggesting that its function may be redundant with that of other sperm proteins.
Collapse
Affiliation(s)
| | - Hamzé JG
- Animal Reproduction Department, INIA-CSIC, Madrid, Spain
- Department of Cell Biology and Histology, Medical School, IMIB, University of Murcia, Murcia, Spain
| | | | - Jiménez-Movilla M
- Department of Cell Biology and Histology, Medical School, IMIB, University of Murcia, Murcia, Spain
- *Correspondence: Jiménez-Movilla M, ; Bermejo-Álvarez P,
| | - Bermejo-Álvarez P
- Animal Reproduction Department, INIA-CSIC, Madrid, Spain
- *Correspondence: Jiménez-Movilla M, ; Bermejo-Álvarez P,
| |
Collapse
|
8
|
Soudy R, Kimura R, Fu W, Patel A, Jhamandas J. Extracellular vesicles enriched with amylin receptor are cytoprotective against the Aß toxicity in vitro. PLoS One 2022; 17:e0267164. [PMID: 35421203 PMCID: PMC9009604 DOI: 10.1371/journal.pone.0267164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/03/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs) are double membrane structures released by all cell types with identified roles in the generation, transportation, and degradation of amyloid-β protein (Aβ) oligomers in Alzheimer’s disease (AD). EVs are thus increasingly recognized to play a neuroprotective role in AD, through their ability to counteract the neurotoxic effects of Aβ, possibly through interactions with specific receptors on cell membranes. Our previous studies have identified the amylin receptor (AMY), particularly AMY3 subtype, as a mediator of the deleterious actions of Aβ in vitro and in vivo experimental paradigms. In the present study, we demonstrate that AMY3 enriched EVs can bind soluble oligomers of Aß and protect N2a cells against toxic effects of this peptide. The effect was specific to amylin receptor as it was blocked in the presence of amylin receptor antagonist AC253. This notion was supported by reduced Aβ binding to EVs from AMY depleted mice compared to those from wild type (Wt) mice. Finally, application of AMY3, but not Wt derived, EVs to hippocampal brain slices improved Aβ-induced reduction of long-term potentiation, a cellular surrogate of memory. Collectively, our observations support the role of AMY receptors, particularly AMY3, in EVs as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ryoichi Kimura
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Center for Liberal Arts and Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jack Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
9
|
Patel A, Kimura R, Fu W, Soudy R, MacTavish D, Westaway D, Yang J, Davey RA, Zajac JD, Jhamandas JH. Genetic Depletion of Amylin/Calcitonin Receptors Improves Memory and Learning in Transgenic Alzheimer's Disease Mouse Models. Mol Neurobiol 2021; 58:5369-5382. [PMID: 34312771 PMCID: PMC8497456 DOI: 10.1007/s12035-021-02490-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023]
Abstract
Based upon its interactions with amyloid β peptide (Aβ), the amylin receptor, a class B G protein-coupled receptor (GPCR), is a potential modulator of Alzheimer's disease (AD) pathogenesis. However, past pharmacological approaches have failed to resolve whether activation or blockade of this receptor would have greater therapeutic benefit. To address this issue, we generated compound mice expressing a human amyloid precursor protein gene with familial AD mutations in combination with deficiency of amylin receptors produced by hemizygosity for the critical calcitonin receptor subunit of this heterodimeric GPCR. These compound transgenic AD mice demonstrated attenuated responses to human amylin- and Aβ-induced depression of hippocampal long-term potentiation (LTP) in keeping with the genetic depletion of amylin receptors. Both the LTP responses and spatial memory (as measured with Morris water maze) in these mice were improved compared to AD mouse controls and, importantly, a reduction in both the amyloid plaque burden and markers of neuroinflammation was observed. Our data support the notion of further development of antagonists of the amylin receptor as AD-modifying therapies.
Collapse
Affiliation(s)
- Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Ryoichi Kimura
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Center for Liberal Arts and Sciences, Sanyo-Onoda City University, Yamaguchi , 756-0884, Japan
| | - Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - David MacTavish
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - David Westaway
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, T6G 2M8, Canada
| | - Jing Yang
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, T6G 2M8, Canada
| | - Rachel A Davey
- Department of Medicine, University of Melbourne, Austin HealthHeidelberg, VIC, 3074, Australia
| | - Jeffrey D Zajac
- Department of Medicine, University of Melbourne, Austin HealthHeidelberg, VIC, 3074, Australia
| | - Jack H Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
10
|
Shin B, Hrdlicka HC, Delany AM, Lee SK. Inhibition of miR-29 Activity in the Myeloid Lineage Increases Response to Calcitonin and Trabecular Bone Volume in Mice. Endocrinology 2021; 162:bqab135. [PMID: 34192317 PMCID: PMC8328098 DOI: 10.1210/endocr/bqab135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 12/29/2022]
Abstract
The miR-29-3p family (miR-29a, miR-29b, miR-29c) of microRNAs is increased during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis. In vivo, activation of a miR-29-3p tough decoy inhibitor in Cre recombinase under the control of the lysozyme 2 promoter-expressing cells (myeloid lineage) resulted in mice displaying enhanced trabecular and cortical bone volume because of decreased bone resorption. Calcitonin receptor (Calcr) is a miR-29 target that negatively regulates bone resorption. CALCR was significantly increased in RANKL-treated miR-29-decoy osteoclasts, and these cells were more responsive to the inhibitory effect of calcitonin on osteoclast formation. Further, cathepsin K (Ctsk), which is critical for resorption, was decreased in miR-29-decoy cells. CALCR is a Gs-coupled receptor and its activation raises cAMP levels. In turn, cAMP suppresses cathepsin K, and cAMP levels were increased in miR-29-decoy cells. siRNA-mediated knock-down of Calcr in miR-29 decoy osteoclasts allowed recovery of cathepsin K levels in these cells. Overall, using a novel knockin tough decoy mouse model, we identified a new role for miR-29-3p in bone homeostasis. In RANKL-driven osteoclastogenesis, as seen in normal bone remodeling, miR-29-3p promotes resorption. Consequently, inhibition of miR-29-3p activity in the myeloid lineage leads to increased trabecular and cortical bone. Further, this study documents an interrelationship between CALCR and CTSK in osteoclastic bone resorption, which is modulated by miR-29-3p.
Collapse
Affiliation(s)
- Bongjin Shin
- Center on Aging, UConn Health, Farmington, CT 06030, USA
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sun-Kyeong Lee
- Center on Aging, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
11
|
Xian Y, Su Y, Liang J, Long F, Feng X, Xiao Y, Lian H, Xu J, Zhao J, Liu Q, Song F. Oroxylin A reduces osteoclast formation and bone resorption via suppressing RANKL-induced ROS and NFATc1 activation. Biochem Pharmacol 2021; 193:114761. [PMID: 34492273 DOI: 10.1016/j.bcp.2021.114761] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 01/15/2023]
Abstract
Excessive bone erosion by osteoclasts is associated with osteoporosis, rheumatoid arthritis, and periprosthetic osteolysis. Targeting osteoclasts may serve as an effective treatment for osteolytic diseases. Although drugs are currently available for the treatment of these diseases, exploring potential anti-osteoclast natural compounds with safe and effective treatment remains needed. Oroxylin A (OA), a natural flavonoid isolated from the root of Scutellaria baicalensis Georgi, has numerous beneficial pharmacological characteristics, including anti-inflammatory and antioxidant activity. However, its effects and mechanisms on osteoclast formation and bone resorption have not yet been clarified. Our research showed that OA attenuated the formation and function of osteoclast induced by RANKL in a time- and concentration-dependent manner without any cytotoxicity. Mechanistically, OA suppressed intracellular reactive oxygen species (ROS) levels through the Nrf2-mediated antioxidant response. Moreover, OA inhibited the activity of NFATc1, the master transcriptional regulator of RANKL-induced osteoclastogenesis. OA exhibited protective effects in mouse models of post-ovariectomy (OVX)- and lipopolysaccharide (LPS)-induced bone loss, in accordance with its in vitro anti-osteoclastogenic effect. Collectively, our findings highlight the potential of OA as a pharmacological agent for the prevention of osteoclast-mediated osteolytic diseases.
Collapse
Affiliation(s)
- Yansi Xian
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiamin Liang
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Feng Long
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoliang Feng
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Xiao
- Medical College of Guangxi University, Nanning, Guangxi, China
| | - Haoyu Lian
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, Australia
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Orthopaedic Department, the First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| | - Fangming Song
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
12
|
Moreira LM, Takawale A, Hulsurkar M, Menassa DA, Antanaviciute A, Lahiri SK, Mehta N, Evans N, Psarros C, Robinson P, Sparrow AJ, Gillis MA, Ashley N, Naud P, Barallobre-Barreiro J, Theofilatos K, Lee A, Norris M, Clarke MV, Russell PK, Casadei B, Bhattacharya S, Zajac JD, Davey RA, Sirois M, Mead A, Simmons A, Mayr M, Sayeed R, Krasopoulos G, Redwood C, Channon KM, Tardif JC, Wehrens XHT, Nattel S, Reilly S. Paracrine signalling by cardiac calcitonin controls atrial fibrogenesis and arrhythmia. Nature 2020; 587:460-465. [PMID: 33149301 DOI: 10.1038/s41586-020-2890-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/13/2020] [Indexed: 11/10/2022]
Abstract
Atrial fibrillation, the most common cardiac arrhythmia, is an important contributor to mortality and morbidity, and particularly to the risk of stroke in humans1. Atrial-tissue fibrosis is a central pathophysiological feature of atrial fibrillation that also hampers its treatment; the underlying molecular mechanisms are poorly understood and warrant investigation given the inadequacy of present therapies2. Here we show that calcitonin, a hormone product of the thyroid gland involved in bone metabolism3, is also produced by atrial cardiomyocytes in substantial quantities and acts as a paracrine signal that affects neighbouring collagen-producing fibroblasts to control their proliferation and secretion of extracellular matrix proteins. Global disruption of calcitonin receptor signalling in mice causes atrial fibrosis and increases susceptibility to atrial fibrillation. In mice in which liver kinase B1 is knocked down specifically in the atria, atrial-specific knockdown of calcitonin promotes atrial fibrosis and increases and prolongs spontaneous episodes of atrial fibrillation, whereas atrial-specific overexpression of calcitonin prevents both atrial fibrosis and fibrillation. Human patients with persistent atrial fibrillation show sixfold lower levels of myocardial calcitonin compared to control individuals with normal heart rhythm, with loss of calcitonin receptors in the fibroblast membrane. Although transcriptome analysis of human atrial fibroblasts reveals little change after exposure to calcitonin, proteomic analysis shows extensive alterations in extracellular matrix proteins and pathways related to fibrogenesis, infection and immune responses, and transcriptional regulation. Strategies to restore disrupted myocardial calcitonin signalling thus may offer therapeutic avenues for patients with atrial fibrillation.
Collapse
Affiliation(s)
- Lucia M Moreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Abhijit Takawale
- Research Centre, Montreal Heart Institute and University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mohit Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - David A Menassa
- Clinical Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Biological Sciences, Faculty of Life and Environmental Sciences, University of Southampton, Southampton, UK
| | - Agne Antanaviciute
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Neelam Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Neil Evans
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Constantinos Psarros
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Alexander J Sparrow
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Marc-Antoine Gillis
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Neil Ashley
- Single-Cell Genomics Facility, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Patrice Naud
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | | | | | - Angela Lee
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Mary Norris
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Michele V Clarke
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Patricia K Russell
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Shoumo Bhattacharya
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Martin Sirois
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Adam Mead
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Alison Simmons
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Rana Sayeed
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford, UK
| | - George Krasopoulos
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford, UK
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jean-Claude Tardif
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Research Centre, Montreal Heart Institute and University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- IHU LIRYC, Fondation Bordeaux Université, Bordeaux, France
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
13
|
Vazifeh Shiran N, Abroun S. Plasma Cell Proliferation Is Reduced in Myeloma-Induced Hypercalcemia and in Co-Culture with Normal Healthy BM-MSCs. Lab Med 2020; 52:273-289. [PMID: 33942854 DOI: 10.1093/labmed/lmaa060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE In multiple myeloma (MM), stimulation of osteoclasts and bone marrow (BM) lesions lead to hypercalcemia, renal failure, and anemia. Co-culture of the myeloma cells in both hypocalcemia and hypercalcemia concentrations with bone marrow-mesenchymal stem cells were evaluated. MATERIALS AND METHODS Viability and survival of myeloma cells were assessed by microculture tetrazolium test and flow cytometric assays. Mesenchymal stem cells (MSCs) were extracted from normal and myeloma patients and were co-cultured with myeloma cells. RESULTS Myeloma cells showed less survival in both hypocalcaemia and hypercalcemia conditions (P <.01). The paracrine and juxtacrine conditions of demineralized bone matrix-induced hypercalcemia increased the proliferation and survival of the cells (P <.05). Unlike myeloma MSCs, normal MSCs reduced the survival of and induced apoptosis in myeloma cells (P <.1). CONCLUSION Normal healthy-MSCs do not protect myeloma cells, but inhibit them. However, increasing the ratio of myeloma cells to MSCs reduces their inhibitory effects of MSCs and leads to their myelomatous transformation.
Collapse
Affiliation(s)
- Nader Vazifeh Shiran
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Abroun
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
14
|
Imprinted genes in clinical exome sequencing: Review of 538 cases and exploration of mouse-human conservation in the identification of novel human disease loci. Eur J Med Genet 2020; 63:103903. [PMID: 32169557 DOI: 10.1016/j.ejmg.2020.103903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 01/20/2020] [Accepted: 03/09/2020] [Indexed: 01/01/2023]
Abstract
Human imprinting disorders cause a range of dysmorphic and neurocognitive phenotypes, and they may elude traditional molecular diagnosis such exome sequencing. The discovery of novel disorders related to imprinted genes has lagged behind traditional Mendelian disorders because current diagnostic technology, especially unbiased testing, has limited utility in their discovery. To identify novel imprinting disorders, we reviewed data for every human gene hypothesized to be imprinted, identified each mouse ortholog, determined its imprinting status in the mouse, and analyzed its function in humans and mice. We identified 17 human genes that are imprinted in both humans and mice, and have functional data in mice or humans to suggest that dysregulated expression would lead to an abnormal phenotype in humans. These 17 genes, along with known imprinted genes, were preferentially flagged 538 clinical exome sequencing tests. The identified genes were: DIRAS3 [1p31.3], TP73 [1p36.32], SLC22A3 [6q25.3], GRB10 [7p12.1], DDC [7p12.2], MAGI2 [7q21.11], PEG10 [7q21.3], PPP1R9A [7q21.3], CALCR [7q21.3], DLGAP2 [8p23.3], GLIS3 [9p24.2], INPP5F [10q26.11], ANO1 [11q13.3], SLC38A4 [12q13.11], GATM [15q21.1], PEG3 [19q13.43], and NLRP2 [19q13.42]. In the 538 clinical cases, eight cases (1.7%) reported variants in a causative known imprinted gene. There were 367/758 variants (48.4%) in imprinted genes that were not known to cause disease, but none of those variants met the criteria for clinical reporting. Imprinted disorders play a significant role in human disease, and additional human imprinted disorders remain to be discovered. Therefore, evolutionary conservation is a potential tool to identify novel genes involved in human imprinting disorders and to identify them in clinical testing.
Collapse
|
15
|
Chakraborty C, Sharma AR, Sharma G, Bhattacharya M, Lee SS. Insight into Evolution and Conservation Patterns of B1-Subfamily Members of GPCR. Int J Pept Res Ther 2020; 26:2505-2517. [PMID: 32421105 PMCID: PMC7223794 DOI: 10.1007/s10989-020-10043-5] [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] [Accepted: 01/30/2020] [Indexed: 11/25/2022]
Abstract
The diverse, evolutionary architectures of proteins can be regarded as molecular fossils, tracing a historical path that marks important milestones across life. The B1-subfamily of GPCRs (G-protein-coupled receptors) are medically significant proteins that comprise 15 transmembrane receptor proteins in Homo sapiens. These proteins control the intracellular concentration of cyclic AMP as well as various vital processes in the body. However, little is known about the evolutionary correlation and conservational blueprint of this GPCR subfamily. We performed a comprehensive analysis to understand the evolutionary architecture among 13 members of the B1-subfamily. Multiple sequence alignment analysis exhibited six multiple sequence aligned blocks and five highly aligned blocks. Molecular phylogenetics indicated that CRHR1 and CRHR2 share a typical ancestral relationship and are siblings in 100% bootstrap replications with a total of 24 nodes observed in the cladogram. CRHR2 has the maximum number of extremely conserved amino acids followed by ADCYAP1R1. The longest continuous number sequence logos (74) were found between sequence location 349 and 423, and consequently, the maximum and minimum logo height recorded was 3.6 bits and 0.18 bits, respectively. Finally, to understand the model and pattern of evolutionary relatedness, the conservation blueprint, and the diversification among the members of a protein family, GPCR distribution from several species throughout the animal kingdom was analysed. Together, the study provides an evolutionary insight and offers a rapid method to explore the potential of depicting the evolutionary relationship, conservation blueprint, and diversification among the B1-subfamily of GPCRs using bioinformatics, algorithm analysis, and mathematical models.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Adamas University, North, 24 Parganas, Kolkata, 700126 West Bengal India
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252 Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252 Republic of Korea
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Manojit Bhattacharya
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252 Republic of Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252 Republic of Korea
| |
Collapse
|
16
|
Yu P, Xie J, Chen Y, Liu J, Liu Y, Bi B, Luo J, Li S, Jiang X, Li J. A thermo-sensitive injectable hydroxypropyl chitin hydrogel for sustained salmon calcitonin release with enhanced osteogenesis and hypocalcemic effects. J Mater Chem B 2020; 8:270-281. [PMID: 31802093 DOI: 10.1039/c9tb02049g] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pharmacotherapy towards hypercalcemia treatment mainly caused by osteoporosis and bone tumor is an effective method to regulate in vivo calcium equilibrium. As a clinical therapeutic peptide, salmon calcitonin (sCT) is considered as a quick-acting medicine but it is limited by the short half-life. To address this challenge, we designed an injectable thermo-sensitive hydrogel based on hydroxypropyl chitin (HPCH) and incorporated the complex of sCT and hyaluronic acid (HA) (sCT-HA) with high association efficiency up to 96.84 ± 7.25%. This composite hydrogel showed a tunable biodegradable property. In vitro sCT release profiles revealed that this hydrogel can achieve long-term sustained sCT release (28 days) with considerable structure stability. The cellular study illustrated outstanding compatibility and osteoconductive potential of this multi-component hydrogel according to the higher ALP activity (2.10-fold), calcium expression (2.30-fold) and extracellular calcium deposition (1.10-fold) compared to that of the sCT group. In vivo sCT release confirmed that this hydrogel system realized sustained sCT release and a continuous hypocalcemic effect for as long as 28 days, and there were no inflammation and immune responses according to the histological evaluations (H&E and IgG staining). These findings demonstrate that this osteoconductive hydrogel system can provide a promising method for therapy of bone related disease.
Collapse
Affiliation(s)
- Peng Yu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Yu Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Jinming Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Yanpeng Liu
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Research Center for Clinical Pharmacy, First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, P. R. China
| | - Bo Bi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Jun Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Sheyu Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| |
Collapse
|
17
|
Xie J, Guo J, Kanwal Z, Wu M, Lv X, Ibrahim NA, Li P, Buabeid MA, Arafa ESA, Sun Q. Calcitonin and Bone Physiology: In Vitro, In Vivo, and Clinical Investigations. Int J Endocrinol 2020; 2020:3236828. [PMID: 32963524 PMCID: PMC7501564 DOI: 10.1155/2020/3236828] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Calcitonin was discovered as a peptide hormone that was known to reduce the calcium levels in the systemic circulation. This hypocalcemic effect is produced due to multiple reasons such as inhibition of bone resorption or suppression of calcium release from the bone. Thus, calcitonin was said as a primary regulator of the bone resorption process. This is the reason why calcitonin has been used widely in clinics for the treatment of bone disorders such as osteoporosis, hypercalcemia, and Paget's disease. However, presently calcitonin usage is declined due to the development of efficacious formulations of new drugs. Calcitonin gene-related peptides and several other peptides such as intermedin, amylin, and adrenomedullin (ADM) are categorized in calcitonin family. These peptides are known for the structural similarity with calcitonin. Aside from having a similar structure, these peptides have few overlapping biological activities and signal transduction action through related receptors. However, several other activities are also present that are peptide specific. In vitro and in vivo studies documented the posttreatment effects of calcitonin peptides, i.e., positive effect on bone osteoblasts and their formation and negative effect on osteoclasts and their resorption. The recent research studies carried out on genetically modified mice showed the inhibition of osteoclast activity by amylin, while astonishingly calcitonin plays its role by suppressing osteoblast and bone turnover. This article describes the review of the bone, the activity of the calcitonin family of peptides, and the link between them.
Collapse
Affiliation(s)
- Jingbo Xie
- Department of Orthopedics, Fengcheng People's Hospital, Fengcheng, Jiangxi 331100, China
| | - Jian Guo
- Department of the Second Orthopedics, Hongdu Hospital of Traditional Chinese Medicine Affiliated to Jiangxi University of Traditional Chinese Medicine, Nanchang Hongdu Traditional Chinese Medicine Hospital, Nanchang, Jiangxi 330008, China
| | | | - Mingzheng Wu
- Department of Orthopaedics, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xiangyang Lv
- Department of Orthopaedics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China
| | | | - Ping Li
- Department of Orthopaedics, Ya'an People's Hospital, Ya'an, Sichuan 625000, China
| | | | | | - Qingshan Sun
- Department of Orthopedics, The Third Hospital of Shandong Province, Jinan, Shandong 250031, China
| |
Collapse
|
18
|
Short amylin receptor antagonist peptides improve memory deficits in Alzheimer's disease mouse model. Sci Rep 2019; 9:10942. [PMID: 31358858 PMCID: PMC6662706 DOI: 10.1038/s41598-019-47255-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022] Open
Abstract
Recent evidence supports involvement of amylin and the amylin receptor in the pathogenesis of Alzheimer’s disease (AD). We have previously shown that amylin receptor antagonist, AC253, improves spatial memory in AD mouse models. Herein, we generated and screened a peptide library and identified two short sequence amylin peptides (12–14 aa) that are proteolytically stable, brain penetrant when administered intraperitoneally, neuroprotective against Aβ toxicity and restore diminished levels of hippocampal long term potentiation in AD mice. Systemic administration of the peptides for five weeks in aged 5XFAD mice improved spatial memory, reduced amyloid plaque burden, and neuroinflammation. The common residue SQELHRLQTY within the peptides is an essential sequence for preservation of the beneficial effects of the fragments that we report here and constitutes a new pharmacological target. These findings suggest that the amylin receptor antagonism may represent a novel therapy for AD.
Collapse
|
19
|
Manglani K, Vijayan V, Pathak C, Khandelwal M, Singh P, Chellappa S, Yadav VK, Surolia A, Gupta S. Development and characterization of supramolecular calcitonin assembly and assessment of its interactions with the bone remodelling process. Bone 2019; 122:123-135. [PMID: 30797058 DOI: 10.1016/j.bone.2019.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
Osteoporosis is the most common metabolic bone disease, which poses an immense socio-economic burden on the society. Human calcitonin, though safe, is not considered as a therapeutic option because of its high tendency to self-associate to form amyloid fibrils thereby affecting its potency. To circumvent this issue we harnessed the inherent capacity of aggregation and developed an assemblage of human calcitonin monomers, [Supramolecular Calcitonin Assembly (SCAI)], which releases biologically active calcitonin monomers in a sustained manner for a period of at least three weeks. AFM and FT-IR analysis showed that SCA-I is amorphous aggregates of calcitonin monomers. Both SCA-I and monomer released from it demonstrated superior anti-osteoclast activity and proteolytic stability in-vitro. SCA-I upon single injection significantly improved bone formation markers and reduced bone resorption markers in ovariectomized (OVX) rat model of postmenopausal osteoporosis. Micro-CT analysis revealed that calcitonin released from SCA-I exhibits its beneficial effect on cortical bone more profoundly compared to trabecular bone. This study demonstrates that SCA-I is more effective compared to the human calcitonin monomers on osteoclasts and has site-specific effect on bone in a model of post-menopausal osteoporosis. This approach opens up an innovative way to use and study the function of human calcitonin.
Collapse
Affiliation(s)
- Kapil Manglani
- Molecular Science Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Viji Vijayan
- Molecular Science Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Chandramani Pathak
- Cell Biology Laboratory, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar 382007, Gujarat, India
| | - Mayuri Khandelwal
- Molecular Science Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Parminder Singh
- Metabolic Research Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Stalin Chellappa
- Molecular Science Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Vijay K Yadav
- Metabolic Research Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Sciences, Bengaluru 560012, Karnataka, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
20
|
Naot D, Musson DS, Cornish J. The Activity of Peptides of the Calcitonin Family in Bone. Physiol Rev 2019; 99:781-805. [PMID: 30540227 DOI: 10.1152/physrev.00066.2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcitonin was discovered over 50 yr ago as a new hormone that rapidly lowers circulating calcium levels. This effect is caused by the inhibition of calcium efflux from bone, as calcitonin is a potent inhibitor of bone resorption. Calcitonin has been in clinical use for conditions of accelerated bone turnover, including Paget's disease and osteoporosis; although in recent years, with the development of drugs that are more potent inhibitors of bone resorption, its use has declined. A number of peptides that are structurally similar to calcitonin form the calcitonin family, which currently includes calcitonin gene-related peptides (αCGRP and βCGRP), amylin, adrenomedullin, and intermedin. Apart from being structurally similar, the peptides signal through related receptors and have some overlapping biological activities, although other activities are peptide specific. In bone, in vitro studies and administration of the peptides to animals generally found inhibitory effects on osteoclasts and bone resorption and positive effects on osteoblasts and bone formation. Surprisingly, studies in genetically modified mice have demonstrated that the physiological role of calcitonin appears to be the inhibition of osteoblast activity and bone turnover, whereas amylin inhibits osteoclast activity. The review article focuses on the activities of peptides of the calcitonin family in bone and the challenges in understanding the relationship between the pharmacological effects and the physiological roles of these peptides.
Collapse
Affiliation(s)
- Dorit Naot
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - David S Musson
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland , Auckland , New Zealand
| |
Collapse
|
21
|
Bartelt A, Jeschke A, Müller B, Gaziano I, Morales M, Yorgan T, Heckt T, Heine M, Gagel RF, Emeson RB, Amling M, Niemeier A, Heeren J, Schinke T, Keller J. Differential effects of Calca-derived peptides in male mice with diet-induced obesity. PLoS One 2017; 12:e0180547. [PMID: 28666011 PMCID: PMC5493411 DOI: 10.1371/journal.pone.0180547] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/16/2017] [Indexed: 12/20/2022] Open
Abstract
Key metabolic hormones, such as insulin, leptin, and adiponectin, have been studied extensively in obesity, however the pathophysiologic relevance of the calcitonin family of peptides remains unclear. This family includes calcitonin (CT), its precursor procalcitonin (PCT), and alpha calcitonin-gene related peptide (αCGRP), which are all encoded by the gene Calca. Here, we studied the role of Calca-derived peptides in diet-induced obesity (DIO) by challenging Calcr-/- (encoding the calcitonin receptor, CTR), Calca-/-, and αCGRP-/- mice and their respective littermates with high-fat diet (HFD) feeding for 16 weeks. HFD-induced pathologies were assessed by glucose tolerance, plasma cytokine and lipid markers, expression studies and histology. We found that DIO in mice lacking the CTR resulted in impaired glucose tolerance, features of enhanced nonalcoholic steatohepatitis (NASH) and adipose tissue inflammation compared to wildtype littermates. Furthermore, CTR-deficient mice were characterized by dyslipidemia and elevated HDL levels. In contrast, mice lacking Calca were protected from DIO, NASH and adipose tissue inflammation, and displayed improved glucose tolerance. Mice exclusively lacking αCGRP displayed a significantly less improved DIO phenotype compared to Calca-deficient mice. In summary, we demonstrate that the CT/CTR axis is involved in regulating plasma cholesterol levels while Calca, presumably through PCT, seems to have a detrimental effect in the context of metabolic disease. Our study provides the first comparative analyses of the roles of Calca-derived peptides and the CTR in metabolic disease.
Collapse
Affiliation(s)
- Alexander Bartelt
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Brigitte Müller
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabella Gaziano
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michelle Morales
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, California, United States of America
| | - Timur Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Heckt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert F. Gagel
- Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ronald B. Emeson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Niemeier
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Keller
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Kim SJ, Ka S, Ha JW, Kim J, Yoo D, Kim K, Lee HK, Lim D, Cho S, Hanotte O, Mwai OA, Dessie T, Kemp S, Oh SJ, Kim H. Cattle genome-wide analysis reveals genetic signatures in trypanotolerant N'Dama. BMC Genomics 2017; 18:371. [PMID: 28499406 PMCID: PMC5427609 DOI: 10.1186/s12864-017-3742-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Indigenous cattle in Africa have adapted to various local environments to acquire superior phenotypes that enhance their survival under harsh conditions. While many studies investigated the adaptation of overall African cattle, genetic characteristics of each breed have been poorly studied. RESULTS We performed the comparative genome-wide analysis to assess evidence for subspeciation within species at the genetic level in trypanotolerant N'Dama cattle. We analysed genetic variation patterns in N'Dama from the genomes of 101 cattle breeds including 48 samples of five indigenous African cattle breeds and 53 samples of various commercial breeds. Analysis of SNP variances between cattle breeds using wMI, XP-CLR, and XP-EHH detected genes containing N'Dama-specific genetic variants and their potential associations. Functional annotation analysis revealed that these genes are associated with ossification, neurological and immune system. Particularly, the genes involved in bone formation indicate that local adaptation of N'Dama may engage in skeletal growth as well as immune systems. CONCLUSIONS Our results imply that N'Dama might have acquired distinct genotypes associated with growth and regulation of regional diseases including trypanosomiasis. Moreover, this study offers significant insights into identifying genetic signatures for natural and artificial selection of diverse African cattle breeds.
Collapse
Affiliation(s)
- Soo-Jin Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - Sojeong Ka
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung-Woo Ha
- Clova, NAVER Corp., Seongnam, 13561, Republic of Korea
| | - Jaemin Kim
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - DongAhn Yoo
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwondo Kim
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hak-Kyo Lee
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, 66414, Republic of Korea
| | - Dajeong Lim
- Division of Animal Genomics and Bioinformatics, National Institute of Animal Science, RDA, Jeonju, 55365, Republic of Korea
| | - Seoae Cho
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - Olivier Hanotte
- University of Nottingham, School of Life Sciences, Nottingham, NG7 2RD, UK.,International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Okeyo Ally Mwai
- International Livestock Research Institute, Box 30709-00100, Nairobi, Kenya
| | - Tadelle Dessie
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Stephen Kemp
- International Livestock Research Institute, Box 30709-00100, Nairobi, Kenya.,The Centre for Tropical Livestock Genetics and Health, The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, Scotland, UK
| | - Sung Jong Oh
- National Institute of Animal Science, RDA, Wanju, 55365, Republic of Korea.
| | - Heebal Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
23
|
Soudy R, Patel A, Fu W, Kaur K, MacTavish D, Westaway D, Davey R, Zajac J, Jhamandas J. Cyclic AC253, a novel amylin receptor antagonist, improves cognitive deficits in a mouse model of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2017; 3:44-56. [PMID: 29067318 PMCID: PMC5651374 DOI: 10.1016/j.trci.2016.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Amylin receptor serves as a portal for the expression of deleterious effects of amyloid β-protein (Aβ), a key pathologic hallmark of Alzheimer's disease. Previously, we showed that AC253, an amylin receptor antagonist, is neuroprotective against Aβ toxicity in vitro and abrogates Aβ-induced impairment of hippocampal long-term potentiation. METHODS Amyloid precursor protein-overexpressing TgCRND8 mice received intracerebroventricularly AC253 for 5 months. New cyclized peptide cAC253 was synthesized and administered intraperitoneally three times a week for 10 weeks in the same mouse model. Cognitive functions were monitored, and pathologic changes were quantified biochemically and immunohistochemically. RESULTS AC253, when administered intracerebroventricularly, improves spatial memory and learning, increases synaptic integrity, reduces microglial activation without discernible adverse effects in TgCRND8 mice. cAC253 demonstrates superior brain permeability, better proteolytic stability, and enhanced binding affinity to brain amylin receptors after a single intraperitoneal injection. Furthermore, cAC253 administered intraperitoneally also demonstrates improvement in spatial memory in TgCRND8 mice. DISCUSSION Amylin receptor is a therapeutic target for Alzheimer's disease and represents a disease-modifying therapy for this condition.
Collapse
Affiliation(s)
- Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kamaljit Kaur
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Chapman University School of Pharmacy, Irvine, California, USA
| | - David MacTavish
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - David Westaway
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel Davey
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Jeffrey Zajac
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Jack Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Kamgar-Parsi K, Tolchard J, Habenstein B, Loquet A, Naito A, Ramamoorthy A. Structural Biology of Calcitonin: From Aqueous Therapeutic Properties to Amyloid Aggregation. Isr J Chem 2016. [DOI: 10.1002/ijch.201600096] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Kian Kamgar-Parsi
- Applied Physics Program; University of Michigan; Ann Arbor MI 48109-1040 USA
| | - James Tolchard
- Institute of Chemistry and Biology of Membranes and Nanoobjects, CNRS, CBMN, UMR 5248; University of Bordeaux; 33600 Pessac France
| | - Birgit Habenstein
- Institute of Chemistry and Biology of Membranes and Nanoobjects, CNRS, CBMN, UMR 5248; University of Bordeaux; 33600 Pessac France
| | - Antoine Loquet
- Institute of Chemistry and Biology of Membranes and Nanoobjects, CNRS, CBMN, UMR 5248; University of Bordeaux; 33600 Pessac France
| | - Akira Naito
- Graduate School of Engineering; Yokohama National University; 79-5 Tokiwadai Hodogaya-ku Yokohama 240-8501 Japan
| | - Ayyalusamy Ramamoorthy
- Department of Chemistry and Biophysics Program; University of Michigan; 930 North University Avenue Ann Arbor MI 48109-1055 USA
| |
Collapse
|
25
|
Henriksen K, Byrjalsen I, Andersen JR, Bihlet AR, Russo LA, Alexandersen P, Valter I, Qvist P, Lau E, Riis BJ, Christiansen C, Karsdal MA. A randomized, double-blind, multicenter, placebo-controlled study to evaluate the efficacy and safety of oral salmon calcitonin in the treatment of osteoporosis in postmenopausal women taking calcium and vitamin D. Bone 2016; 91:122-9. [PMID: 27462009 DOI: 10.1016/j.bone.2016.07.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/11/2016] [Accepted: 07/22/2016] [Indexed: 01/09/2023]
Abstract
This randomized, double-blind, placebo-controlled phase III study was conducted to assess the efficacy and safety of oral calcitonin (SMC021) for the treatment of postmenopausal osteoporosis. A total of 4665 postmenopausal women with osteoporosis were randomized 1:1 to receive calcium and vitamin D plus either SMC021 tablets (0.8mg/d) or placebo for 36months. The primary endpoint was the proportion of patients with a new vertebral fracture. The two groups were well balanced at baseline with regards to demographic and clinical data. No effect of SMC021 on preventing new vertebral fractures was observed, nor was any effect seen on new hip or non-vertebral fractures. Women receiving SMC021 had a mean 1.02% (±0.12%) increase in lumbar spine bone mineral density (BMD) compared with a mean 0.18% (±0.12%) increase in the placebo group by the end of the study (p<0.0001). Similarly, small increases in BMD were observed at the femoral neck and hip in both groups. Levels of the biomarkers of bone turnover, urinary CTX-I and CTX-II, were 15% lower in the SMC021 group than in the placebo arm at 12 and 24months, but not at 36months. No change in quality of life between groups, assessed by the Qualeffo-14 questionnaire, was observed in either group between baseline and month 36. Pharmacokinetics analysis confirmed exposure to SMC021, but the drug levels were markedly lower than expected. Approximately 92% of subjects in each treatment group experienced an adverse event (AE), the majority of which were mild or moderate in intensity. AEs associated with SMC021 were primarily of gastrointestinal origin and included nausea, vomiting and abdominal pain, as well as hot flushes which were the reason for the slightly higher drop-out rate in the active treatment arm compared to placebo. The number of severe AEs was low in both groups. Thirty-five deaths were reported but none were considered treatment-related. Due to the lack of efficacy in preventing fractures, the development of the orally formulated calcitonin was terminated despite the promising results in earlier studies.
Collapse
Affiliation(s)
- Kim Henriksen
- Nordic Bioscience Biomarkers & Research, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark.
| | - Inger Byrjalsen
- Nordic Bioscience Clinical Development, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Jeppe R Andersen
- Nordic Bioscience Clinical Development, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Asger R Bihlet
- Nordic Bioscience Clinical Development, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Luis A Russo
- Center for Clinical and Basic Research (CCBR), Rio de Janeiro, Brazil
| | | | - Ivo Valter
- Center for Clinical and Basic Research (CCBR), Tallinn, Estonia
| | - Per Qvist
- Nordic Bioscience Biomarkers & Research, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Edith Lau
- Center for Clinical and Basic Research (CCBR), Hong Kong, China
| | - Bente J Riis
- Nordic Bioscience Clinical Development, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Claus Christiansen
- Nordic Bioscience Clinical Development, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| | - Morten A Karsdal
- Nordic Bioscience Biomarkers & Research, Nordic Bioscience A/S, Herlev Hovedgade 205-207, DK-2730 Herlev, Denmark
| |
Collapse
|
26
|
Śliwiński L, Cegieła U, Pytlik M, Folwarczna J, Janas A, Zbrojkiewicz M. Effects of fenoterol on the skeletal system depend on the androgen level. Pharmacol Rep 2016; 69:260-267. [PMID: 28126642 DOI: 10.1016/j.pharep.2016.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/22/2016] [Accepted: 09/28/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND The role of sympathetic nervous system in the osseous tissue remodeling is not clear enough. METHODS The effects of fenoterol, a selective β2-adrenomimetic drug, on the skeletal system of normal and androgen deficient (orchidectomized) rats were studied in vivo. Osteoclastogenesis and mRNA expression in osteoblasts were investigated in vitro in mouse cell cultures. RESULTS Fenoterol administered to animals with physiological androgen level unfavorably affected the skeletal system, damaging the bone microarchitecture. Androgen deficiency induced osteoporotic changes, and fenoterol protected the osseous tissue from consequences of androgen deficiency. The results of in vitro studies correlated with the in vivo observations. A significantly increased number of osteoclasts in bone marrow cell cultures to which testosterone and fenoterol were added simultaneously was demonstrated. In cultures without the addition of testosterone, fenoterol significantly inhibited osteoclastogenesis in comparison with control cultures. CONCLUSIONS The results indicate the favorable action of fenoterol in conditions of testosterone deficiency, and its destructive influence upon the skeleton in the presence of androgens. The results confirm the key role of sympathetic nervous system in the regulation of bone remodeling.
Collapse
Affiliation(s)
- Leszek Śliwiński
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland.
| | - Urszula Cegieła
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Maria Pytlik
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Joanna Folwarczna
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Aleksandra Janas
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Małgorzata Zbrojkiewicz
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
27
|
Kovacs CS. Maternal Mineral and Bone Metabolism During Pregnancy, Lactation, and Post-Weaning Recovery. Physiol Rev 2016; 96:449-547. [PMID: 26887676 DOI: 10.1152/physrev.00027.2015] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During pregnancy and lactation, female physiology adapts to meet the added nutritional demands of fetuses and neonates. An average full-term fetus contains ∼30 g calcium, 20 g phosphorus, and 0.8 g magnesium. About 80% of mineral is accreted during the third trimester; calcium transfers at 300-350 mg/day during the final 6 wk. The neonate requires 200 mg calcium daily from milk during the first 6 mo, and 120 mg calcium from milk during the second 6 mo (additional calcium comes from solid foods). Calcium transfers can be more than double and triple these values, respectively, in women who nurse twins and triplets. About 25% of dietary calcium is normally absorbed in healthy adults. Average maternal calcium intakes in American and Canadian women are insufficient to meet the fetal and neonatal calcium requirements if normal efficiency of intestinal calcium absorption is relied upon. However, several adaptations are invoked to meet the fetal and neonatal demands for mineral without requiring increased intakes by the mother. During pregnancy the efficiency of intestinal calcium absorption doubles, whereas during lactation the maternal skeleton is resorbed to provide calcium for milk. This review addresses our current knowledge regarding maternal adaptations in mineral and skeletal homeostasis that occur during pregnancy, lactation, and post-weaning recovery. Also considered are the impacts that these adaptations have on biochemical and hormonal parameters of mineral homeostasis, the consequences for long-term skeletal health, and the presentation and management of disorders of mineral and bone metabolism.
Collapse
Affiliation(s)
- Christopher S Kovacs
- Faculty of Medicine-Endocrinology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
28
|
Tsukamoto M, Menuki K, Murai T, Hatakeyama A, Takada S, Furukawa K, Sakai A. Elcatonin prevents bone loss caused by skeletal unloading by inhibiting preosteoclast fusion through the unloading-induced high expression of calcitonin receptors in bone marrow cells. Bone 2016; 85:70-80. [PMID: 26851124 DOI: 10.1016/j.bone.2016.01.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 11/20/2022]
Abstract
This study aimed to clarify whether elcatonin (EL) has a preventive action on bone dynamics in skeletal unloading. Seven-week-old male C57BL/6J mice with either ground control (GC) or tail suspension (TS) were administered EL 20U/kg or a vehicle (veh) three times per week and assigned to one of the following four groups: GCEL, GCveh, TSEL, and TSveh. Blood samples and bilateral femurs and tibias of the mice were obtained for analysis. After 7days of unloading, the trabecular bone mineral density in the distal femur obtained via peripheral quantitative computed tomography and the trabecular bone volume were significantly higher in the TSEL group than in the TSveh group. The bone resorption histomorphometric parameters, such as the osteoclast surface and osteoclast number, were significantly suppressed in the TSEL mice, whereas the number of preosteoclasts was significantly increased. The plasma level of tartrate-resistant acid phosphatase-5b (TRACP-5b) was significantly lower in the TSEL group than in all other groups. In the bone marrow cell culture, the number of TRACP-positive (TRACP(+)) multinucleated cells was significantly lower in the TSEL mice than in the TSveh mice, whereas the number of TRACP(+) mononucleated cells was higher in the TSEL mice. On day 4, the expression of nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1 (NFATc1), cathepsin K and d2 isoform of vacuolar ATPase V0 domain (ATP6V0D2) mRNA in the bone marrow cells in the TSEL mice was suppressed, and the expression of calcitonin receptor (Calcr) mRNA on day 1 and Calcr antigen on day 4 were significantly higher in the TSveh mice than in the GCveh mice. EL prevented the unloading-induced bone loss associated with the high expression of Calcr in the bone marrow cells of mouse hindlimbs after tail suspension, and it suppressed osteoclast development from preosteoclasts to mature osteoclasts through bone-resorbing activity. This study of EL-treated unloaded mice provides the first in vivo evidence of a physiological role of EL in the inhibition of the differentiation process from preosteoclasts to osteoclasts.
Collapse
Affiliation(s)
- Manabu Tsukamoto
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Kunitaka Menuki
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Teppei Murai
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Akihisa Hatakeyama
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Shinichiro Takada
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Kayoko Furukawa
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Akinori Sakai
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
29
|
Clarke MV, Russell PK, Findlay DM, Sastra S, Anderson PH, Skinner JP, Atkins GJ, Zajac JD, Davey RA. A Role for the Calcitonin Receptor to Limit Bone Loss During Lactation in Female Mice by Inhibiting Osteocytic Osteolysis. Endocrinology 2015; 156:3203-14. [PMID: 26135836 DOI: 10.1210/en.2015-1345] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During lactation, the large transfer of calcium from the mother to the milk is primarily sourced from the maternal skeleton. To determine whether the calcitonin receptor (CTR) plays a physiological role to protect the skeleton from excessive resorption during lactation, we assessed the maternal skeleton of global CTR knockout (CTRKO) and littermate control mice at the end of lactation (postnatal day 21). Micro-computed tomography analyses showed no effect on trabecular or cortical bone in the distal femur and L1 vertebra of maternal global CTR deletion at the end of lactation in global CTRKO mice compared with that in control mice. Bone resorption, as assessed by osteoclast number and activity at the end of lactation, was unaffected by maternal CTR deletion. Cathepsin K, carbonic anhydrase 2, matrix metalloproteinase 13, and receptor activator of nuclear factor-κB ligand mRNA levels, however, were markedly elevated by 3- to 6.5-fold in whole bone of lactating global CTRKO females. Because these genes have been shown to be up-regulated in osteocytes during lactation when osteocytes resorb their surrounding bone matrix, together with their reported expression of the CTR, we determined the osteocyte lacunar area in cortical bone. After lactation, the top 20% of osteocyte lacunar area in global CTRKO mice was 10% larger than the top 20% in control mice. These data are consistent with an increased osteocytic osteolysis in global CTRKO mice during lactation, which is further supported by the increased serum calcium observed in global CTRKO mice after lactation. These results provide evidence for a physiological role for the CTR to protect the maternal skeleton during lactation by a direct action on osteocytes to inhibit osteolysis.
Collapse
Affiliation(s)
- Michele V Clarke
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Patricia K Russell
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - David M Findlay
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Stephen Sastra
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Paul H Anderson
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Jarrod P Skinner
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Gerald J Atkins
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Jeffrey D Zajac
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| | - Rachel A Davey
- Department of Medicine (M.V.C., P.K.R., S.S., J.P.S., J.D.Z., R.A.D.), Austin Health, University of Melbourne, Heidelberg, 3084 Victoria, Australia; Centre for Orthopaedic and Trauma Research (D.M.F., G.J.A.), University of Adelaide, 5005 Adelaide, Australia; and School of Pharmacy and Medical Sciences (P.H.A.), University of South Australia, 5005 Adelaide, Australia
| |
Collapse
|
30
|
Hay DL, Chen S, Lutz TA, Parkes DG, Roth JD. Amylin: Pharmacology, Physiology, and Clinical Potential. Pharmacol Rev 2015; 67:564-600. [PMID: 26071095 DOI: 10.1124/pr.115.010629] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amylin is a pancreatic β-cell hormone that produces effects in several different organ systems. Here, we review the literature in rodents and in humans on amylin research since its discovery as a hormone about 25 years ago. Amylin is a 37-amino-acid peptide that activates its specific receptors, which are multisubunit G protein-coupled receptors resulting from the coexpression of a core receptor protein with receptor activity-modifying proteins, resulting in multiple receptor subtypes. Amylin's major role is as a glucoregulatory hormone, and it is an important regulator of energy metabolism in health and disease. Other amylin actions have also been reported, such as on the cardiovascular system or on bone. Amylin acts principally in the circumventricular organs of the central nervous system and functionally interacts with other metabolically active hormones such as cholecystokinin, leptin, and estradiol. The amylin-based peptide, pramlintide, is used clinically to treat type 1 and type 2 diabetes. Clinical studies in obesity have shown that amylin agonists could also be useful for weight loss, especially in combination with other agents.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Steve Chen
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Thomas A Lutz
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - David G Parkes
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Jonathan D Roth
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| |
Collapse
|
31
|
Russell PK, Clarke MV, Cheong K, Anderson PH, Morris HA, Wiren KM, Zajac JD, Davey RA. Androgen receptor action in osteoblasts in male mice is dependent on their stage of maturation. J Bone Miner Res 2015; 30:809-23. [PMID: 25407961 DOI: 10.1002/jbmr.2413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 11/10/2022]
Abstract
Androgen action via the androgen receptor (AR) is essential for normal skeletal growth and bone maintenance post-puberty in males; however, the molecular and cellular mechanisms by which androgens exert their actions in osteoblasts remains relatively unexplored in vivo. To identify autonomous AR actions in osteoblasts independent of AR signaling in other tissues, we compared the extent to which the bone phenotype of the Global-ARKO mouse was restored by replacing the AR in osteoblasts commencing at either the (1) proliferative or (2) mineralization stage of their maturation. In trabecular bone, androgens stimulated trabecular bone accrual during growth via the AR in proliferating osteoblasts and maintained trabecular bone post-puberty via the AR in mineralizing osteoblasts, with its predominant action being to inhibit bone resorption by decreasing the ratio of receptor activator of NF-κB ligand (RANKL) to osteoprotegerin (OPG) gene expression. During growth, replacement of the AR in proliferating but not mineralizing osteoblasts of Global-ARKOs was able to partially restore periosteal circumference, supporting the concept that androgen action in cortical bone to increase bone size during growth is mediated via the AR in proliferating osteoblasts. This study provides further significant insight into the mechanism of androgen action via the AR in osteoblasts, demonstrating that it is dependent on the stage of osteoblast maturation.
Collapse
Affiliation(s)
- Patricia K Russell
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Australia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Felsenfeld AJ, Levine BS. Calcitonin, the forgotten hormone: does it deserve to be forgotten? Clin Kidney J 2015; 8:180-7. [PMID: 25815174 PMCID: PMC4370311 DOI: 10.1093/ckj/sfv011] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 01/30/2015] [Indexed: 12/12/2022] Open
Abstract
Calcitonin is a 32 amino acid hormone secreted by the C-cells of the thyroid gland. Calcitonin has been preserved during the transition from ocean-based life to land dwellers and is phylogenetically older than parathyroid hormone. Calcitonin secretion is stimulated by increases in the serum calcium concentration and calcitonin protects against the development of hypercalcemia. Calcitonin is also stimulated by gastrointestinal hormones such as gastrin. This has led to the unproven hypothesis that postprandial calcitonin stimulation could play a role in the deposition of calcium and phosphate in bone after feeding. However, no bone or other abnormalities have been described in states of calcitonin deficiency or excess except for diarrhea in a few patients with medullary thyroid carcinoma. Calcitonin is known to stimulate renal 1,25 (OH)2 vitamin D (1,25D) production at a site in the proximal tubule different from parathyroid hormone and hypophosphatemia. During pregnancy and lactation, both calcitonin and 1,25D are increased. The increases in calcitonin and 1,25D may be important in the transfer of maternal calcium to the fetus/infant and in the prevention and recovery of maternal bone loss. Calcitonin has an immediate effect on decreasing osteoclast activity and has been used for treatment of hypercalcemia. Recent studies in the calcitonin gene knockout mouse have shown increases in bone mass and bone formation. This last result together with the presence of calcitonin receptors on the osteocyte suggests that calcitonin could possibly affect osteocyte products which affect bone formation. In summary, a precise role for calcitonin remains elusive more than 50 years after its discovery.
Collapse
Affiliation(s)
- Arnold J Felsenfeld
- Department of Medicine , VA Greater Los Angeles Healthcare System and the David Geffen School of Medicine at UCLA , Los Angeles, CA , USA
| | - Barton S Levine
- Department of Medicine , VA Greater Los Angeles Healthcare System and the David Geffen School of Medicine at UCLA , Los Angeles, CA , USA
| |
Collapse
|
33
|
The role of calcitonin receptor signalling in polyethylene particle-induced osteolysis. Acta Biomater 2015; 14:125-32. [PMID: 25486133 DOI: 10.1016/j.actbio.2014.11.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 11/14/2014] [Accepted: 11/28/2014] [Indexed: 12/28/2022]
Abstract
The detection of peptides from the calcitonin (CT) family in the periarticular tissue of loosened implants has raised hopes of opening new regenerative therapies in the process of aseptic loosening, which remains the major cause of early implant failure in endoprosthetic surgery. We have previously shown the roles of α-calcitonin gene-related peptide (α-CGRP) and the CALCA gene which encodes α-CGRP/CT in this process. To uncover the role of direct calcitonin receptor (CTR) mediated signalling, we studied particle-induced osteolysis (PIO) in a murine calvaria model with a global deletion of the CTR (CTR-KO) using μCT analysis and histomorphometry. As expected, CTR-KO mice revealed reduced bone volume compared to wild-type (WT) controls (p<0.05). In CTR-KO mice we found significantly higher RANKL (receptor activator of NF-κB ligand) expression in the particle group than in the control group. The increase in osteoclast numbers by the particles was twice as high as the increase of osteoclasts in the WT mice (400 vs. 200%). Changes in the eroded surface and actual osteolysis due to ultrahigh-molecular-weight polyethylene particles were similar in WTs and CTR-KOs. Taken together, our findings strengthen the relevance of the OPG/RANK/RANKL system in the PIO process. CTR seems to have an effect on osteoclast differentiation in this context. As there were no obvious changes of the amount of PIO in CTR deficiency, regenerative strategies in aseptic loosening of endoprosthetic implants based on peptides arising from the CT family should rather focus on the impact of α-CGRP.
Collapse
|
34
|
Wang H, Zhuo Y, Hu X, Shen W, Zhang Y, Chu T. CD147 deficiency blocks IL-8 secretion and inhibits lung cancer-induced osteoclastogenesis. Biochem Biophys Res Commun 2015; 458:268-73. [DOI: 10.1016/j.bbrc.2015.01.097] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/21/2015] [Indexed: 12/28/2022]
|
35
|
Martin TJ, Sims NA. Calcitonin physiology, saved by a lysophospholipid. J Bone Miner Res 2015; 30:212-5. [PMID: 25581311 DOI: 10.1002/jbmr.2449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/24/2022]
Affiliation(s)
- T John Martin
- St. Vincent's Institute of Medical Research and The University of Melbourne, Department of Medicine, St. Vincent's Hospital, Fitzroy, Australia
| | | |
Collapse
|
36
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
37
|
Calcitonin controls bone formation by inhibiting the release of sphingosine 1-phosphate from osteoclasts. Nat Commun 2014; 5:5215. [PMID: 25333900 PMCID: PMC4205484 DOI: 10.1038/ncomms6215] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022] Open
Abstract
The hormone calcitonin (CT) is primarily known for its pharmacologic
action as an inhibitor of bone resorption, yet CT-deficient mice display increased bone formation. These findings
raised the question about the underlying cellular and molecular mechanism of
CT action. Here we show that either
ubiquitous or osteoclast-specific inactivation of the murine CT receptor (CTR) causes increased bone formation. CT negatively regulates the osteoclast expression
of Spns2 gene, which encodes a
transporter for the signalling lipid sphingosine
1-phosphate (S1P).
CTR-deficient mice show increased
S1P levels, and their skeletal
phenotype is normalized by deletion of the S1P receptor S1P3. Finally, pharmacologic treatment
with the nonselective S1P receptor agonist FTY720 causes increased bone formation in wild-type, but not in
S1P3-deficient mice.
This study redefines the role of CT in
skeletal biology, confirms that S1P
acts as an osteoanabolic molecule in vivo and provides evidence for a
pharmacologically exploitable crosstalk between osteoclasts and osteoblasts. The regulatory role of calcitonin in bone homeostasis is well studied,
yet its molecular activity is poorly understood. The authors show that calcitonin regulates
bone cells function by inhibiting the osteoclast secretion of sphingosine 1-phosphate, a
lipid mediator of osteoclast–osteoblast crosstalk.
Collapse
|
38
|
Qiu WQ, Zhu H. Amylin and its analogs: a friend or foe for the treatment of Alzheimer's disease? Front Aging Neurosci 2014; 6:186. [PMID: 25120481 PMCID: PMC4114192 DOI: 10.3389/fnagi.2014.00186] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/09/2014] [Indexed: 12/20/2022] Open
Abstract
Amylin, a gut-brain axis hormone, and amyloid-beta peptides (Aβ), a major component of the Alzheimer's disease (AD) brain, share several features, including similar β-sheet secondary structures, binding to the same receptor and being degraded by the same protease, insulin degrading enzyme (IDE). However, while amylin readily crosses the blood brain barrier (BBB) and mediates several activities including improving glucose metabolism, relaxing cerebrovascular structure, modulating inflammatory reaction and perhaps enhancing neural regeneration, Aβ has no known physiological functions. Thus, abundant Aβ in the AD brain could block or interfere with the binding of amylin to its receptor and hinder its functions. Recent studies using animal models for AD demonstrate that amylin and its analog reduce the AD pathology in the brain and improve cognitive impairment in AD. Given that, in addition to amyloid plaques and neurofibrillary tangles, perturbed cerebral glucose metabolism and cerebrovascular damage are the hallmarks of the AD brain, we propose that giving exogenous amylin type peptides have the potential to become a new avenue for the diagnosis and therapeutic of AD. Although amylin's property of self-aggregation may be a limitation to developing it as a therapeutic for AD, its clinical analog, pramlintide containing 3 amino acid differences from amylin, does not aggregate like human amylin, but more potently mediates amylin's activities in the brain. Pramlintide is an effective drug for diabetes with a favorable profile of safety. Thus a randomized, double-blind, placebo-controlled clinical trial should be conducted to examine the efficacy of pramlintide for AD. This review summarizes the knowledge and findings on amylin type peptides and discuss pros and cons for their potential for AD.
Collapse
Affiliation(s)
- Wei Qiao Qiu
- Department of Psychiatry, Boston University School of Medicine Boston, MA, USA ; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine Boston, MA, USA ; Alzheimer's Disease Center, Boston University School of Medicine Boston, MA, USA
| | - Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine Boston, MA, USA
| |
Collapse
|
39
|
Association of calcitonin receptor gene polymorphism with bone mineral density in postmenopausal Chinese women: a meta-analysis. Arch Gynecol Obstet 2014; 291:165-72. [PMID: 25055932 DOI: 10.1007/s00404-014-3378-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 07/09/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The relationship between Calcitonin receptor (CTR) gene polymorphism C1377T and bone mineral density (BMD) in postmenopausal women has been studied to some degree in Western countries, but the reports remain inconclusive and have not been generalized to other populations. PURPOSE To evaluate the association of CTR gene polymorphism C1377T with BMD in the Han Chinese population. METHODS We searched for all published articles indexed in MEDLINE (1950-2014), EMBASE (1974-2014), China National Knowledge Infrastructure (CNKI, 1994-2014), and the Wanfang Database, using the key words "polymorphism," "CTR," "osteoporosis," and "bone density." The data were extracted independently by two reviewers. The heterogeneity between studies was determined using the Chi-square-based Q test. Potential publication bias was estimated using a funnel plot and Egger's linear regression test. Odds ratios and 95 % confidence intervals (CI) were used to evaluate the results. RESULTS Six eligible studies were selected for the meta-analysis. Our analysis suggested that the C1377T polymorphism of the CTR gene was associated with BMD at the lumbar spine (95 % CI -0.57 to -0.05; P = 0.02), but not associated with BMD at the femoral neck (95 % CI -0.27 to 0.24; P = 0.90) in the postmenopausal Han Chinese population. CONCLUSION The C1377T polymorphism in the CTR gene is associated with BMD at the lumbar spine in a postmenopausal Han Chinese population and the CTR gene may become a useful genetic marker for predicting the risk of developing osteoporosis in postmenopausal Chinese women.
Collapse
|
40
|
Kovacs CS. The Role of PTHrP in Regulating Mineral Metabolism During Pregnancy, Lactation, and Fetal/Neonatal Development. Clin Rev Bone Miner Metab 2014. [DOI: 10.1007/s12018-014-9157-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
How JMY, Wardak SA, Ameer SI, Davey RA, Sartor DM. Blunted sympathoinhibitory responses in obesity-related hypertension are due to aberrant central but not peripheral signalling mechanisms. J Physiol 2014; 592:1705-20. [PMID: 24492842 DOI: 10.1113/jphysiol.2013.269670] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The gut hormone cholecystokinin (CCK) acts at subdiaphragmatic vagal afferents to induce renal and splanchnic sympathoinhibition and vasodilatation, via reflex inhibition of a subclass of cardiovascular-controlling neurons in the rostroventrolateral medulla (RVLM). These sympathoinhibitory and vasodilator responses are blunted in obese, hypertensive rats and our aim in the present study was to determine whether this is attributable to (i) altered sensitivity of presympathetic vasomotor RVLM neurons, and (ii) aberrant peripheral or central signalling mechanisms. Using a diet-induced obesity model, male Sprague-Dawley rats exhibited either an obesity-prone (OP) or obesity-resistant (OR) phenotype when placed on a medium high fat diet for 13-15 weeks; control animals were placed on a low fat diet. OP animals had elevated resting arterial pressure compared to OR/control animals (P < 0.05). Barosensitivity of RVLM neurons was significantly attenuated in OP animals (P < 0.05), suggesting altered baroreflex gain. CCK induced inhibitory responses in RVLM neurons of OR/control animals but not OP animals. Subdiaphragmatic vagal nerve responsiveness to CCK and CCK1 receptor mRNA expression in nodose ganglia did not differ between the groups, but CCK induced significantly less Fos-like immunoreactivity in both the nucleus of the solitary tract and the caudal ventrolateral medulla of OP animals compared to controls (P < 0.05). These results suggest that blunted sympathoinhibitory and vasodilator responses in obesity-related hypertension are due to alterations in RVLM neuronal responses, resulting from aberrant central but not peripheral signalling mechanisms. In obesity, blunted sympathoinhibitory mechanisms may lead to increased regional vascular resistance and contribute to the development of hypertension.
Collapse
|
42
|
Ell B, Mercatali L, Ibrahim T, Campbell N, Schwarzenbach H, Pantel K, Amadori D, Kang Y. Tumor-induced osteoclast miRNA changes as regulators and biomarkers of osteolytic bone metastasis. Cancer Cell 2013; 24:542-56. [PMID: 24135284 PMCID: PMC3832956 DOI: 10.1016/j.ccr.2013.09.008] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 07/15/2013] [Accepted: 09/13/2013] [Indexed: 02/06/2023]
Abstract
Understanding the mechanism by which tumor cells influence osteoclast differentiation is crucial for improving treatment of osteolytic metastasis. Here, we report broad microRNA (miRNA) expression changes in differentiating osteoclasts after exposure to tumor-conditioned media, in part through activation of NFκB signaling by soluble intracellular adhesion molecule (sICAM1) secreted from bone-metastatic cancer cells. Ectopic expression of multiple miRNAs downregulated during osteoclastogenesis suppresses osteoclast differentiation by targeting important osteoclast genes. Intravenous delivery of these miRNAs in vivo inhibits osteoclast activity and reduces osteolytic bone metastasis. Importantly, serum levels of sICAM1 and two osteoclast miRNAs, miR-16 and miR-378, which are elevated in osteoclast differentiation, correlate with bone metastasis burden. These findings establish miRNAs as potential therapeutic targets and clinical biomarkers of bone metastasis.
Collapse
Affiliation(s)
- Brian Ell
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Mabilleau G, Mieczkowska A, Irwin N, Flatt PR, Chappard D. Optimal bone mechanical and material properties require a functional glucagon-like peptide-1 receptor. J Endocrinol 2013; 219:59-68. [PMID: 23911987 DOI: 10.1530/joe-13-0146] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone is permanently remodeled by a complex network of local, hormonal, and neuronal factors that affect osteoclast and osteoblast biology. Among these factors, a role for gastrointestinal hormones has been proposed based on the evidence that bone resorption dramatically falls after a meal. Glucagon-like peptide-1 (GLP1) is one of these gut hormones, and despite several reports suggesting an anabolic effect of GLP1, or its stable analogs, on bone mass, little is known about the effects of GLP1/GLP1 receptor on bone strength. In this study, we investigated by three-point bending, quantitative X-ray microradiography, microcomputed tomography, qBEI, and FTIRI bone strength and bone quality in male Glp1r knockout (Glp1r KO) mice when compared with control WT animals. Animals with a deletion of Glp1r presented with a significant reduction in ultimate load, yield load, stiffness, and total absorbed and post-yield energies when compared with WT animals. Furthermore, cortical thickness and bone outer diameter were significantly decreased in deficient animals. The mineral quantity and quality were not significantly different between Glp1r KO and WT animals. On the other hand, the maturity of the collagen matrix was significantly reduced in deficient animals and associated with lowered material properties. Taken together, these data support a positive effect of GLP1R on bone strength and quality.
Collapse
Affiliation(s)
- Guillaume Mabilleau
- Groupe d'Etudes sur le Remodelage Osseux et les bioMatériaux (GEROM)-LHEA, Service Commun d'Imageries et d'Analyses Microscopiques (SCIAM), IRIS-IBS Institut de Biologie en Santé, LUNAM Université, CHU d'Angers, 49933 Angers Cedex, France
| | | | | | | | | |
Collapse
|
44
|
Conigrave AD, Ward DT. Calcium-sensing receptor (CaSR): pharmacological properties and signaling pathways. Best Pract Res Clin Endocrinol Metab 2013; 27:315-31. [PMID: 23856262 DOI: 10.1016/j.beem.2013.05.010] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In this article we consider the mechanisms by which the calcium-sensing receptor (CaSR) induces its cellular responses via the control (activation or inhibition) of signaling pathways. We consider key features of CaSR-mediated signaling including its control of the heterotrimeric G-proteins Gq/11, Gi/o and G12/13 and the downstream consequences recognizing that very few CaSR-mediated cell phenomena have been fully described. We also consider the manner in which the CaSR contributes to the formation of specific signaling scaffolds via peptide recognition sequences in its intracellular C-terminal along with the origins of its high level of cooperativity, particularly for Ca(2+)o, and its remarkable resistance to desensitization. We also consider the nature of the mechanisms by which the CaSR controls oscillatory and sustained Ca(2+)i mobilizing responses and inhibits or elevates cyclic adenosine monophosphate (cAMP) levels dependent on the cellular and signaling context. Finally, we consider the diversity of the receptor's ligands, ligand binding sites and broader compartment-dependent physiological roles leading to the identification of pronounced ligand-biased signaling for agonists including Sr(2+) and modulators including l-amino acids and the clinically effective calcimimetic cinacalcet. We note the implications of these findings for the development of new designer drugs that might target the CaSR in pathophysiological contexts beyond those established for the treatment of disorders of calcium metabolism.
Collapse
Affiliation(s)
- Arthur D Conigrave
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia.
| | | |
Collapse
|
45
|
Davey RA, Findlay DM. Calcitonin: physiology or fantasy? J Bone Miner Res 2013; 28:973-9. [PMID: 23519892 DOI: 10.1002/jbmr.1869] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 12/13/2012] [Accepted: 01/03/2013] [Indexed: 01/04/2023]
Abstract
Calcitonin, a potent hypocalcemic hormone produced by the C-cells of the thyroid, was first discovered by Harold Copp in 1962. The physiological significance of calcitonin has been questioned, but recent studies using genetically modified mouse models have uncovered additional actions of calcitonin acting through its receptor (CTR) that are of particular significance to the regulation of bone and calcium homeostasis. Mice in which the CTR is deleted in osteoclasts are more susceptible to induced hypercalcemia and exogenous calcitonin is able to lower serum calcium in younger animals. These data are consistent with the hypothesis that calcitonin can regulate serum calcium by inhibiting the efflux of calcium from bone, and that this action is most important when bone turnover is high. Calcitonin has also been implicated in protecting the skeleton from excessive loss of bone mineral during times of high calcium demand, such as lactation. This action may be linked to an intriguing and as yet unexplained observation that calcitonin inhibits bone formation, because deletion of the CTR leads to increased bone formation. We propose several mechanisms by which calcitonin could protect the skeleton by regulating bone turnover, acting within the bone and/or centrally. A new more holistic notion of the physiological role of calcitonin in bone and calcium homeostasis is required and we have highlighted some important knowledge gaps so that future calcitonin research will help to achieve such an understanding.
Collapse
Affiliation(s)
- Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.
| | | |
Collapse
|
46
|
Cinacalcet attenuates hypercalcemia observed in mice bearing either Rice H-500 Leydig cell or C26-DCT colon tumors. Eur J Pharmacol 2013; 712:8-15. [PMID: 23623934 DOI: 10.1016/j.ejphar.2013.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 03/27/2013] [Accepted: 04/03/2013] [Indexed: 11/22/2022]
Abstract
Excessive secretion of parathyroid hormone-related protein (PTHrP) by tumors stimulates bone resorption and increases renal tubular reabsorption of calcium, resulting in hypercalcemia of malignancy. We investigated the ability of cinacalcet, an allosteric modulator of the calcium-sensing receptor, to attenuate hypercalcemia by assessing its effects on blood ionized calcium, serum PTHrP, and calcium-sensing receptor mRNA in mice bearing either Rice H-500 Leydig cell or C26-DCT colon tumors. Cinacalcet effectively decreased hypercalcemia in a dose- and enantiomer-dependent manner; furthermore, cinacalcet normalized phosphorus levels, but did not affect serum PTHrP. Ribonuclease protection assay results demonstrated presence of PTHrP receptor, but not calcium-sensing receptor mRNA in C26-DCT tumors. The mechanism by which cinacalcet lowered serum calcium was investigated in parathyroidectomized rats (i.e., without PTH) made hypercalcemic by PTHrP. Cinacalcet attenuated PTHrP-mediated elevations in blood ionized calcium, which were accompanied by increased plasma calcitonin. Taken together these results suggest that the cinacalcet-mediated decrease in serum calcium is not the result of a direct effect on tumor cells, but rather is the result of increased calcitonin release. In summary, cinacalcet effectively reduced tumor-mediated hypercalcemia and corrected hypophosphatemia in mice. Further investigation of cinacalcet for treatment of hypercalcemia of malignancy is warranted.
Collapse
|
47
|
Roth JD. Amylin and the regulation of appetite and adiposity: recent advances in receptor signaling, neurobiology and pharmacology. Curr Opin Endocrinol Diabetes Obes 2013. [PMID: 23183359 DOI: 10.1097/med.0b013e32835b896f] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW This review focuses on recent advances in receptor signaling, neurobiology, and pharmacological interactions of amylin with nutritive status, as well as other metabolism-related regulatory signals. RECENT FINDINGS Manipulation of components of the amylin receptor complex revealed important roles for the accessory proteins of amylin receptors in energy balance. In-vitro findings point to potential novel sites of action and postreceptor signaling pathways activated by amylin. Neurobiological studies elucidated how amylin activation of hindbrain neural circuitry modulates hypothalamic signaling and responsiveness to leptin. The notion of 'amylin resistance' was addressed in several models (drug or diet-induced hyper-amylinemia). Finally, progress in the design and delivery of amylinomimetics is briefly discussed. SUMMARY Collectively, these mechanistic studies deepen our understanding of the role of endogenous amylin in the regulation of appetite and adiposity, and hopefully will help guide research efforts towards the development of more effective amylin-based therapies for metabolic diseases.
Collapse
|
48
|
Roth JD, Erickson MR, Chen S, Parkes DG. GLP-1R and amylin agonism in metabolic disease: complementary mechanisms and future opportunities. Br J Pharmacol 2012; 166:121-36. [PMID: 21671898 DOI: 10.1111/j.1476-5381.2011.01537.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The discoveries of the incretin hormone glucagon-like peptide-1 (GLP-1) and the β-cell hormone amylin have translated into hormone-based therapies for diabetes. Both classes of molecules also exhibit weight-lowering effects and have been investigated for their anti-obesity potential. In the present review, we explore the mechanisms underlying the physiological and pharmacological actions of GLP-1 and amylin agonism. Despite their similarities (e.g. both molecular classes slow gastric emptying, decrease glucagon and inhibit food intake), there are important distinctions between the central and/or peripheral pathways that mediate their effects on glycaemia and energy balance. We suggest that understanding the similarities and differences between these molecules holds important implications for the development of novel, combination-based therapies, which are increasingly the norm for diabetes/metabolic disease. Finally, the future of GLP-1- and amylin agonist-based therapeutics is discussed.
Collapse
|
49
|
Kuo YJ, Tsuang FY, Sun JS, Lin CH, Chen CH, Li JY, Huang YC, Chen WY, Yeh CB, Shyu JF. Calcitonin inhibits SDCP-induced osteoclast apoptosis and increases its efficacy in a rat model of osteoporosis. PLoS One 2012; 7:e40272. [PMID: 22792258 PMCID: PMC3391248 DOI: 10.1371/journal.pone.0040272] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/04/2012] [Indexed: 12/13/2022] Open
Abstract
Introduction Treatment for osteoporosis commonly includes the use of bisphosphonates. Serious side effects of these drugs are caused by the inhibition of bone resorption as a result of osteoclast apoptosis. Treatment using calcitonin along with bisphosphonates overcomes these side-effects in some patients. Calcitonin is known to inhibit bone resorption without reducing the number of osteoclasts and is thought to prolong osteoclast survival through the inhibition of apoptosis. Further understanding of how calcitonin inhibits apoptosis could prove useful to the development of alternative treatment regimens for osteoporosis. This study aimed to analyze the mechanism by which calcitonin influences osteoclast apoptosis induced by a bisphosphate analog, sintered dicalcium pyrophosphate (SDCP), and to determine the effects of co-treatment with calcitonin and SDCP on apoptotic signaling in osteoclasts. Methods Isolated osteoclasts were treated with CT, SDCP or both for 48 h. Osteoclast apoptosis assays, pit formation assays, and tartrate-resistant acid phosphatase (TRAP) staining were performed. Using an osteoporosis rat model, ovariectomized (OVX) rats received calcitonin, SDCP, or calcitonin + SDCP. The microarchitecture of the fifth lumbar trabecular bone was investigated, and histomorphometric and biochemical analyses were performed. Results Calcitonin inhibited SDCP-induced apoptosis in primary osteoclast cultures, increased Bcl-2 and Erk activity, and decreased Mcl-1 activity. Calcitonin prevented decreased osteoclast survival but not resorption induced by SDCP. Histomorphometric analysis of the tibia revealed increased bone formation, and microcomputed tomography of the fifth lumbar vertebrate showed an additive effect of calcitonin and SDCP on bone volume. Finally, analysis of the serum bone markers CTX-I and P1NP suggests that the increased bone volume induced by co-treatment with calcitonin and SDCP may be due to decreased bone resorption and increased bone formation. Conclusions Calcitonin reduces SDCP-induced osteoclast apoptosis and increases its efficacy in an in vivo model of osteoporosis.
Collapse
Affiliation(s)
- Yi-Jie Kuo
- Department of Orthopaedic, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan, Republic of China
| | - Fon-Yih Tsuang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Jui-Sheng Sun
- Department of Orthopaedic Surgery, National Taiwan University Hospital-Hsin Chu, Hsin-Chu, Taiwan, Republic of China
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chi-Hung Lin
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei, Taiwan, Republic of China
| | - Chia-Hsien Chen
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei, Taiwan, Republic of China
| | - Jia-Ying Li
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yi-Chian Huang
- Institute of Anatomy and Cell Biology National Yang Ming University, Taipei, Taiwan, Republic of China
| | - Wei-Yu Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chin-Bin Yeh
- Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Jia-Fwu Shyu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
50
|
Davey RA, Clarke MV, Sastra S, Skinner JP, Chiang C, Anderson PH, Zajac JD. Decreased body weight in young Osterix-Cre transgenic mice results in delayed cortical bone expansion and accrual. Transgenic Res 2011; 21:885-93. [PMID: 22160436 DOI: 10.1007/s11248-011-9581-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 11/30/2011] [Indexed: 12/18/2022]
Abstract
Conditional gene inactivation using the Cre/loxP system has lead to significant advances in our understanding of the function of genes in a wide range of disciplines. It is becoming increasingly apparent in the literature, that Cre transgenic mice may themselves have a phenotype. In the following study we describe the bone phenotype of a commonly used Cre transgenic mouse line to study osteoblasts, the Osx-GFP::Cre (Osx-Cre) mice. Cortical and trabecular bone parameters were determined in the femurs of Osx-Cre mice at 6 and 12 weeks of age by microtomography (μCT). At 6 weeks of age, Osx-Cre mice had reduced body weight by 22% (P < 0.0001) and delayed cortical bone expansion and accrual, characterized by decreases in periosteal circumference by 7% (P < 0.05) and cortical thickness by 11% (P < 0.01), compared to wild type controls. Importantly, the cortical bone phenotype of the skeletally immature Osx-Cre mice at 6 weeks of age could be accounted for by their low body weight. The delayed weight gain and cortical growth of Osx-Cre mice was overcome by 12 weeks of age, with no differences observed between Osx-Cre and wild type controls. In conclusion, Osx-Cre expressing mice display a delayed growth phenotype in the absence of doxycycline treatment, evidenced by decreased cortical bone expansion and accrual at 6 weeks of age, as an indirect result of decreased body weight. While this delay in growth is overcome by adulthood at 12 weeks of age, caution together with appropriate data analysis must be considered when assessing the experimental data from skeletally immature Cre/loxP knockout mice generated using the Osx-Cre mouse line to avoid misinterpretation.
Collapse
Affiliation(s)
- Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Studley Road, Heidelberg, VIC 3084, Australia.
| | | | | | | | | | | | | |
Collapse
|