1
|
Xu D, Wu H, Zhou C. Fusion of parathyroid hormone (1–34) to an albumin-binding domain improves osteogenesis. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
2
|
Xiong Y, Hirano H, Lane NE, Nandi S, McDonald KA. Plant-based production and characterization of a promising Fc-fusion protein against microgravity-induced bone density loss. Front Bioeng Biotechnol 2022; 10:962292. [PMID: 36172011 PMCID: PMC9511166 DOI: 10.3389/fbioe.2022.962292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Microgravity-induced bone loss is a main obstacle for long term space missions as it is difficult to maintain bone mass when loading stimuli is reduced. With a typical bone mineral density loss of 1.5% per month of microgravity exposure, the chances for osteoporosis and fractures may endanger astronauts’ health. Parathyroid Hormone or PTH (1–34) is an FDA approved treatment for osteoporosis, and may reverse microgravity-induced bone loss. However, PTH proteins requires refrigeration, daily subcutaneous injection, and have a short shelf-life, limiting its use in a resource-limited environment, like space. In this study, PTH was produced in an Fc-fusion form via transient expression in plants, to improve the circulatory half-life which reduces dosing frequency and to simplify purification if needed. Plant-based expression is well-suited for space medicine application given its low resource consumption and short expression timeline. The PTH-Fc accumulation profile in plant was established with a peak expression on day 5 post infiltration of 373 ± 59 mg/kg leaf fresh weight. Once the PTH-Fc was purified, the amino acid sequence and the binding affinity to its target, PTH 1 receptor (PTH1R), was determined utilizing biolayer interferometry (BLI). The binding affinity between PTH-Fc and PTH1R was 2.30 × 10−6 M, similar to the affinity between PTH (1–34) and PTH1R (2.31 × 10−6 M). Its function was also confirmed in a cell-based receptor stimulation assay, where PTH-Fc was able to stimulate the PTH1R producing cyclic adenosine monophosphate (cAMP) with an EC50 of (8.54 ± 0.12) x 10−9 M, comparable to the EC50 from the PTH (1–34) of 1.49 × 10−8 M. These results suggest that plant recombinant PTH-Fc exhibits a similar binding affinity and potency in a PTH1R activation assay compared to PTH. Furthermore, it can be produced rapidly at high levels with minimal resources and reagents, making it ideal for production in low resource environments such as space.
Collapse
Affiliation(s)
- Yongao Xiong
- Department of Chemical Engineering, University of California, Davis, CA, United States
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
| | - Hiroto Hirano
- Department of Chemical Engineering, University of California, Davis, CA, United States
| | - Nancy E. Lane
- Center for Musculoskeletal Health, School of Medicine, University of California, Davis, CA, United States
| | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, CA, United States
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Global HealthShare, University of California, Davis, CA, United States
| | - Karen A. McDonald
- Department of Chemical Engineering, University of California, Davis, CA, United States
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, United States
- Global HealthShare, University of California, Davis, CA, United States
- *Correspondence: Karen A. McDonald,
| |
Collapse
|
3
|
Rana D, Salave S, Jain S, Shah R, Benival D. Systematic Development and Optimization of Teriparatide-Loaded Nanoliposomes Employing Quality by Design Approach for Osteoporosis. J Pharm Innov 2022. [DOI: 10.1007/s12247-022-09663-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
4
|
Yu J, Song Y, Yang A, Zhang X, Li L. Serum nuclear factor IB as a novel and noninvasive indicator in the diagnosis of secondary hyperparathyroidism. J Clin Lab Anal 2021; 35:e23787. [PMID: 33991027 PMCID: PMC8183937 DOI: 10.1002/jcla.23787] [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: 01/11/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Chronic renal failure (CRF) referred to chronic progressive renal parenchymal damage caused by various causes, with metabolite retention and imbalance of water, electrolyte, and acid-base balance as the main clinical manifestations. Secondary hyperparathyroidism (sHPT) was a common complication in maintenance hemodialysis patients with CRF. Nuclear factor IB (NFIB) was a newly found tumor suppressor gene in various cancers. The present study aimed to illustrate the role of NFIB in sHPT clinical diagnosis and treatment response. METHODS A retrospective, case-control study, including 189 patients with sHPT and 106 CRF patients without sHPT, compared with 95 controls. Serum NFIB and 1,25(OH)2 D3 levels were measured by RT-qPCR and ELISAs, respectively. ROC analysis was conducted to verify the diagnostic value of NFIB in sHPT. Spearman's correlation analysis was conducted to verify the association between NFIB and bone mineral density (BMD) scores. After 6 months of treatment, the variance of NFIB and 1,25(OH)2 D3 in different groups was recorded. RESULTS The expression of NFIB was significantly lower in serum samples from sHPT and non-sHPT CRF patients, compared to controls. Clinicopathological information verified sHPT was associated with NFIB, parathyroid hormone (PTH), serum calcium, serum phosphorus, time of dialysis, and serum 1,25(OH)2 D3 levels. Spearman's correlation analysis illustrated the positive correlation between NFIB levels and BMD scores. At receiver operator characteristic (ROC) curve analysis, the cutoff of 1.6508 for NFIB was able to identify patients with sHPT from healthy controls; meanwhile, NFIB could also discriminate sHPT among CRF patients as well (cutoff = 1.4741). Furthermore, we found that during 6 months of treatment, NFIB levels were gradually increased, while PTH and serum P levels were decreased. CONCLUSIONS Serum NFIB was a highly accurate tool to identify sHPT from healthy controls and CRF patients. Due to its simplicity, specificity, and sensitivity, this candidate can be proposed as a first-line examination in the diagnostic workup in sHPT.
Collapse
Affiliation(s)
- Jian'gen Yu
- Department of Nephrology, The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Yu Song
- Department of Nephrology, The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Aihua Yang
- Department of Nephrology, The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Xiaoyun Zhang
- Department of Nephrology, The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Lin Li
- Department of Nephrology, The First People's Hospital of Xiaoshan District, Hangzhou, China
| |
Collapse
|
5
|
Yin L, Zhu Y, He M, Chang Y, Xu F, Lai H. Preparation and characteristics of electrospinning PTH‐Fc/PLCL/SF membranes for bioengineering applications. J Biomed Mater Res A 2019; 108:157-165. [PMID: 31566865 DOI: 10.1002/jbm.a.36801] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/22/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Lihua Yin
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of StomatologyShanghai JiaoTong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology Shanghai China
- Department of ImplantologySchool/Hospital of Stomatology Lanzhou University Lanzhou Gansu China
| | - Yidan Zhu
- Department of Stomatology Technology, School of MedicineXi'an International University Xi'an Shaanxi China
| | - Miaomiao He
- Department of VIP Dental Care Center of Hangzhou West Dental Hospital Zhejiang Hangzhou China
| | - Yaoren Chang
- Department of ImplantologySchool/Hospital of Stomatology Lanzhou University Lanzhou Gansu China
| | - Fangfang Xu
- Department of ImplantologySchool/Hospital of Stomatology Lanzhou University Lanzhou Gansu China
| | - Hong‐Chang Lai
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of StomatologyShanghai JiaoTong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology Shanghai China
| |
Collapse
|
6
|
Prospects of Parathyroid Hormone in Therapeutic Intervention. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
7
|
van Gastel J, Hendrickx JO, Leysen H, Santos-Otte P, Luttrell LM, Martin B, Maudsley S. β-Arrestin Based Receptor Signaling Paradigms: Potential Therapeutic Targets for Complex Age-Related Disorders. Front Pharmacol 2018; 9:1369. [PMID: 30546309 PMCID: PMC6280185 DOI: 10.3389/fphar.2018.01369] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
G protein coupled receptors (GPCRs) were first characterized as signal transducers that elicit downstream effects through modulation of guanine (G) nucleotide-binding proteins. The pharmacotherapeutic exploitation of this signaling paradigm has created a drug-based field covering nearly 50% of the current pharmacopeia. Since the groundbreaking discoveries of the late 1990s to the present day, it is now clear however that GPCRs can also generate productive signaling cascades through the modulation of β-arrestin functionality. β-Arrestins were first thought to only regulate receptor desensitization and internalization - exemplified by the action of visual arrestin with respect to rhodopsin desensitization. Nearly 20 years ago, it was found that rather than controlling GPCR signal termination, productive β-arrestin dependent GPCR signaling paradigms were highly dependent on multi-protein complex formation and generated long-lasting cellular effects, in contrast to G protein signaling which is transient and functions through soluble second messenger systems. β-Arrestin signaling was then first shown to activate mitogen activated protein kinase signaling in a G protein-independent manner and eventually initiate protein transcription - thus controlling expression patterns of downstream proteins. While the possibility of developing β-arrestin biased or functionally selective ligands is now being investigated, no additional research has been performed on its possible contextual specificity in treating age-related disorders. The ability of β-arrestin-dependent signaling to control complex and multidimensional protein expression patterns makes this therapeutic strategy feasible, as treating complex age-related disorders will likely require therapeutics that can exert network-level efficacy profiles. It is our understanding that therapeutically targeting G protein-independent effectors such as β-arrestin will aid in the development of precision medicines with tailored efficacy profiles for disease/age-specific contextualities.
Collapse
Affiliation(s)
- Jaana van Gastel
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Jhana O Hendrickx
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Hanne Leysen
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt University of Berlin, Berlin, Germany
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes and Medical Genetics, Medical University of South Carolina, Charleston, SC, United States
| | - Bronwen Martin
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| |
Collapse
|
8
|
|
9
|
Miller PD, Pannacciulli N, Brown JP, Czerwinski E, Nedergaard BS, Bolognese MA, Malouf J, Bone HG, Reginster JY, Singer A, Wang C, Wagman RB, Cummings SR. Denosumab or Zoledronic Acid in Postmenopausal Women With Osteoporosis Previously Treated With Oral Bisphosphonates. J Clin Endocrinol Metab 2016; 101:3163-70. [PMID: 27270237 PMCID: PMC4971333 DOI: 10.1210/jc.2016-1801] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CONTEXT Denosumab and zoledronic acid (ZOL) are parenteral treatments for patients with osteoporosis. OBJECTIVE The objective of the study was to compare the effect of transitioning from oral bisphosphonates to denosumab or ZOL on bone mineral density (BMD) and bone turnover. DESIGN AND SETTING This was an international, multicenter, randomized, double-blind trial. PARTICIPANTS A total of 643 postmenopausal women with osteoporosis previously treated with oral bisphosphonates participated in the study. INTERVENTIONS Subjects were randomized 1:1 to sc denosumab 60 mg every 6 months plus iv placebo once or ZOL 5 mg iv once plus sc placebo every 6 months for 12 months. MAIN OUTCOME MEASURES Changes in BMD and bone turnover markers were measured. RESULTS BMD change from baseline at month 12 was significantly greater with denosumab compared with ZOL at the lumbar spine (primary end point; 3.2% vs 1.1%; P < .0001), total hip (1.9% vs 0.6%; P < .0001), femoral neck (1.2% vs -0.1%; P < .0001), and one-third radius (0.6% vs 0.0%; P < .05). The median decrease from baseline was greater with denosumab than ZOL for serum C-telopeptide of type 1 collagen at all time points after day 10 and for serum procollagen type 1 N-terminal propeptide at month 1 and at all time points after month 3 (all P < .05). Median percentage changes from baseline in serum intact PTH were significantly greater at months 3 and 9 with denosumab compared with ZOL (all P < .05). Adverse events were similar between groups. Three events consistent with the definition of atypical femoral fracture were observed (two denosumab and one ZOL). CONCLUSIONS In postmenopausal women with osteoporosis previously treated with oral bisphosphonates, denosumab was associated with greater BMD increases at all measured skeletal sites and greater inhibition of bone remodeling compared with ZOL.
Collapse
Affiliation(s)
- P D Miller
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - N Pannacciulli
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - J P Brown
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - E Czerwinski
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - B S Nedergaard
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - M A Bolognese
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - J Malouf
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - H G Bone
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - J-Y Reginster
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - A Singer
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - C Wang
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - R B Wagman
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| | - S R Cummings
- Colorado Center for Bone Research (P.D.M.), Lakewood, Colorado 80277; Amgen Inc (N.P., C.W., R.B.W.), Thousand Oaks, California 91320; Laval University and Centre Hospitalier Universitaire de Québec Research Centre (J.P.B.), Québec City, Québec G1V 4G2, Canada; Krakow Medical Center (E.C.), 31-501 Krakow, Poland; Center for Clinical and Basic Research (B.S.N.), Aalborg, DK-9000 Aalborg, Denmark; Bethesda Health Research Center (M.A.B.), Bethesda, Maryland 20817; Hospital de la Santa Creu i Sant Pau (J.M.), 08025 Barcelona, Spain; Michigan Bone and Mineral Clinic (H.G.B.), Detroit, Michigan 48236; University of Liège (J.-Y.R.), 4000 Liège, Belgium; Georgetown University Medical Center (A.S.), Washington, DC 20007; and San Francisco Coordinating Center (S.R.C.), California Pacific Medical Center Research Institute, San Francisco, California 94143
| |
Collapse
|
10
|
Abstract
Osteoporosis is a chronic disease of the osseous system characterized by decreased bone strength and increased fracture risk. It is due to an imbalance in the dynamic ongoing processes of bone formation and bone resorption. Currently available osteoporosis therapies like bisphosphonates, selective estrogen receptor modulators (SERMs), and denosumab are anti-resorptive agents. Parathyroid hormone analogs like teriparatide are the only anabolic agents currently approved for osteoporosis treatment. The side-effects and limited efficacy of the presently available therapies has encouraged extensive research into the pathophysiology of the disease and newer drug targets for its treatment. The novel anti-resorptive agents being developed are newer SERMs, osteoprotegerin, c-src (cellular-sarcoma) kinase inhibitors, αVβ3 integrin antagonists, cathepsin K inhibitors, chloride channel inhibitors, and nitrates. Upcoming anabolic agents include calcilytics, antibodies against sclerostin and Dickkopf-1, statins, matrix extracellular phosphoglycoprotein fragments activin inhibitiors, and endo-cannabinoid agonists. Many of these new drugs are still in development. This article provides an insight into the emerging drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Garima Bhutani
- Department of Pharmacology, Pt. B.D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| | - Mahesh Chander Gupta
- Department of Pharmacology, Pt. B.D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| |
Collapse
|
11
|
Cheloha RW, Maeda A, Dean T, Gardella TJ, Gellman SH. Backbone modification of a polypeptide drug alters duration of action in vivo. Nat Biotechnol 2014; 32:653-5. [PMID: 24929976 DOI: 10.1038/nbt.2920] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 05/05/2014] [Indexed: 11/09/2022]
Abstract
Systematic modification of the backbone of bioactive polypeptides through β-amino acid residue incorporation could provide a strategy for generating molecules with improved drug properties, but such alterations can result in lower receptor affinity and potency. Using an agonist of parathyroid hormone receptor-1 (PTHR1), a G protein-coupled receptor in the B-family, we present an approach for α→β residue replacement that enables both high activity and improved pharmacokinetic properties in vivo.
Collapse
Affiliation(s)
- Ross W Cheloha
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Akira Maeda
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Dean
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel H Gellman
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Stratford R, Vu C, Sakon J, Katikaneni R, Gensure R, Ponnapakkam T. Pharmacokinetics in rats of a long-acting human parathyroid hormone-collagen binding domain peptide construct. J Pharm Sci 2014; 103:768-75. [PMID: 24399637 DOI: 10.1002/jps.23843] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 12/13/2013] [Accepted: 12/13/2013] [Indexed: 01/16/2023]
Abstract
The pharmacokinetics of a hybrid peptide consisting of the N-terminal biologically active region of human parathyroid hormone (PTH) linked to a collagen-binding domain (CBD) were evaluated in female Sprague-Dawley rats. The peptide, PTH-CBD, consists of the first 33 amino acids of PTH linked as an extension of the amino acid chain to the CBD peptide derived from ColH collagenase of Clostridium histolyticum. Serum concentrations arising from single dose administration by the subcutaneous and intravenous routes were compared with those measured following route-specific mole equivalent doses of PTH(1-34). Population-based modeling demonstrated similar systemic absorption kinetics and bioavailability for both peptides. Exposure to PTH-CBD was sixfold higher because of a systemic clearance of approximately 20% relative to PTH(1-34); however, these kinetics were consistent with more than 95% of a dose being eliminated from serum within 24 h. Results obtained support continued investigation of PTH-CBD as a bone-targeted anabolic agent for the treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Robert Stratford
- College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana, 70130
| | | | | | | | | | | |
Collapse
|
13
|
Ponnapakkam T, Katikaneni R, Sakon J, Stratford R, Gensure RC. Treating osteoporosis by targeting parathyroid hormone to bone. Drug Discov Today 2013; 19:204-8. [PMID: 23932952 DOI: 10.1016/j.drudis.2013.07.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/19/2013] [Accepted: 07/25/2013] [Indexed: 01/24/2023]
Abstract
Osteoporosis is a major public health problem despite widespread use of bisphosphonate therapy. PTH(1-34) is a more effective treatment; but its use has been limited by side effects (hypercalcemia, tumor risk) and inconvenient dosing (daily injection). Long-acting forms of PTH are also effective but cause severe hypercalcemia, presumably from effects in kidney. We hypothesized that targeted delivery of PTH to bone using a collagen binding domain (PTH-CBD) could reduce hypercalcemia. PTH-CBD is cleared from serum within 12hours after subcutaneous administration. In ovariectomized rats, monthly administration of PTH-CBD increased spinal BMD by 14.2% with no associated hypercalcemia. Such bone-targeted anabolic agents may ultimately allow the superior efficacy of anabolic therapy to be obtained with the dosing convenience of bisphosphonates.
Collapse
Affiliation(s)
- T Ponnapakkam
- Pediatric Endocrinology, Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA.
| | - R Katikaneni
- Pediatric Endocrinology, Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Sakon
- Chemistry, University of Arkansas, Fayetteville, AR, USA
| | - R Stratford
- College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, USA
| | - R C Gensure
- Pediatric Endocrinology, Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
14
|
Smith EL, Kanczler JM, Roberts CA, Oreffo ROC. Developmental cues for bone formation from parathyroid hormone and parathyroid hormone-related protein in an ex vivo organotypic culture system of embryonic chick femora. Tissue Eng Part C Methods 2012; 18:984-94. [PMID: 22690868 PMCID: PMC4014091 DOI: 10.1089/ten.tec.2012.0132] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 06/04/2012] [Indexed: 11/13/2022] Open
Abstract
Enhancement and application of our understanding of skeletal developmental biology is critical to developing tissue engineering approaches to bone repair. We propose that use of the developing embryonic femur as a model to further understand skeletogenesis, and the effects of key differentiation agents, will aid our understanding of the developing bone niche and inform bone reparation. We have used a three-dimensional organotypic culture system of embryonic chick femora to investigate the effects of two key skeletal differentiation agents, parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP), on bone and cartilage development, using a combination of microcomputed tomography and histological analysis to assess tissue formation and structure, and cellular behavior. Stimulation of embryonic day 11 (E11) organotypic femur cultures with PTH and PTHrP initiated osteogenesis. Bone formation was enhanced, with increased collagen I and STRO-1 expression, and cartilage was reduced, with decreased chondrocyte proliferation, collagen II expression, and glycosaminoglycan levels. This study demonstrates the successful use of organotypic chick femur cultures as a model for bone development, evidenced by the ability of exogenous bioactive molecules to differentially modulate bone and cartilage formation. The organotypic model outlined provides a tool for analyzing key temporal stages of bone and cartilage development, providing a paradigm for translation of bone development to improve scaffolds and skeletal stem cell treatments for skeletal regenerative medicine.
Collapse
Affiliation(s)
- Emma L Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton Medical School, Southampton, United Kingdom.
| | | | | | | |
Collapse
|
15
|
Ponnapakkam T, Katikaneni R, Suda H, Miyata S, Matsushita O, Sakon J, Gensure RC. A single injection of the anabolic bone agent, parathyroid hormone-collagen binding domain (PTH-CBD), results in sustained increases in bone mineral density for up to 12 months in normal female mice. Calcif Tissue Int 2012; 91:196-203. [PMID: 22806683 PMCID: PMC3693552 DOI: 10.1007/s00223-012-9626-1] [Citation(s) in RCA: 18] [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: 12/14/2011] [Accepted: 05/29/2012] [Indexed: 01/08/2023]
Abstract
Parathyroid hormone (PTH) is the most effective osteoporosis treatment, but it is only effective if administered by daily injections. We fused PTH(1-33) to a collagen binding domain (PTH-CBD) to extend its activity, and have shown an anabolic bone effect with monthly dosing. We tested the duration of action of this compound with different routes of administration. Normal young C57BL/6J mice received a single intraperitoneal injection of PTH-CBD (320 μg/kg). PTH-CBD treated mice showed a 22.2 % increase in bone mineral density (BMD) at 6 months and 12.8 % increase at 12 months. When administered by subcutaneous injection, PTH-CBD again caused increases in BMD, 15.2 % at 6 months and 14.3 % at 12 months. Radiolabeled PTH-CBD was concentrated in bone and skin after either route of administration. We further investigated skin effects of PTH-CBD, and histological analysis revealed an apparent increase in anagen VI hair follicles. A single dose of PTH-CBD caused sustained increases in BMD by >10 % for 1 year in normal mice, regardless of the route of administration, thus showing promise as a potential osteoporosis therapy.
Collapse
Affiliation(s)
- Tulasi Ponnapakkam
- Department of Pediatric Endocrinology, Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
16
|
Matsumoto T, Kuriwaka-Kido R, Kondo T, Endo I, Kido S. Regulation of osteoblast differentiation by interleukin-11 via AP-1 and Smad signaling. Endocr J 2012; 59:91-101. [PMID: 21931225 DOI: 10.1507/endocrj.ej11-0219] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Mechanical stress and parathyroid hormone (PTH) are major stimulators, and aging and glucocorticoids excess are important suppressors of osteoblast differentiation. Mechanical stress and PTH stimulate interleukin (IL)-11 expression in cells of osteoblast lineage by enhancing transcription of IL-11 gene via an increase in intracellular Ca²⁺. The elevated Ca²⁺ activates extracellular signal-regulated kinase (ERK) to enhance phosphorylation of cyclic AMP response element-binding protein (CREB), which binds to the fosB gene promoter and enhances ΔFosB expression. ΔFosB dimerizes with JunD on the IL-11 gene promoter to enhance its transcription. Both mechanical stress and PTH also stimulate phosphorylation of Smad1 via an activation of protein kinase Cδ (PKCδ). Phosphorylated Smad1 binds to the IL-11 gene promoter and forms complex with ΔFosB/JunD to further enhance IL-11 gene transcription. The increased IL-11 then suppresses expression of Wnt inhibitors, including Dickkopf 1 (Dkk1) and 2, and enhances Wnt signaling to stimulate osteoblast differentiation and inhibit adipocyte differentiation. The suppression of osteoblast differentiation by aging involves a decrease in IL-11 gene transcription by a reduction in JunD binding to the activator protein (AP)-1 site of the IL-11 gene promoter. Glucocorticoids inhibit transcriptional activation of IL-11 gene by an interaction of glucocorticoid-glucocorticoid receptor (GR) complex with ΔFosB/JunD heterodimer. Thus, factors that enhance osteoblast differentiation stimulate, and those which suppress osteoblast differentiation inhibit IL-11 gene transcription, and IL-11 enhances Wnt signaling by suppressing expression of its inhibitors. These observations are consistent with the notion that IL-11 mediates stimulatory and inhibitory signals of osteoblast differentiation by affecting Wnt signaling.
Collapse
Affiliation(s)
- Toshio Matsumoto
- Department of Medicine and Bioregulatory Sciences, The University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan.
| | | | | | | | | |
Collapse
|
17
|
Ponnapakkam T, Katikaneni R, Miller E, Ponnapakkam A, Hirofumi S, Miyata S, Suva LJ, Sakon J, Matsushita O, Gensure RC. Monthly administration of a novel PTH-collagen binding domain fusion protein is anabolic in mice. Calcif Tissue Int 2011; 88:511-20. [PMID: 21512758 DOI: 10.1007/s00223-011-9485-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 03/22/2011] [Indexed: 10/18/2022]
Abstract
We synthesized fusion proteins of parathyroid hormone (PTH) (1-33) and the collagen binding domain of ColH (CBD) and tested them for anabolic bone activity in mice. Two fusion proteins were synthesized, linking the carboxy terminus of PTH(1-33) either directly to the amino terminal of the CBD or to the CBD through an adjacent ColH domain (PTH-PKD-CBD). Both PTH-CBD and PTH-PKD-CBD increased cAMP accumulation in cells stably transfected with the PTH/PTHrP receptor, and both peptides bound to type 1 collagen in flow-through assays. Distribution studies indicated that the PTH-CBD was concentrated in the bone and skin, tissues with abundant collagen and blood flow. Administration of 320 μg/kg PTH-CBD either weekly (for 8 weeks) or monthly (for 6 months) to 7-week-old C57BL/6J mice resulted in a sustained increase in bone mineral density (BMD) (15% for weekly studies, 13% for monthly studies; P < 0.05). PTH-PKD-CBD showed only 5% increases in BMD after weekly administration, and, as expected, neither weekly nor monthly PTH(1-34) affected BMD. PTH-CBD increased serum alkaline phosphatase levels. Importantly, there were no significant increases in serum calcium observed. Collectively, the data suggest that PTH-CBD has a sustained anabolic effect in bone with either weekly or monthly administration. This approach of targeted delivery of PTH to bone may show promise for the treatment of disorders of low bone mass, such as postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Tulasi Ponnapakkam
- Pediatric Endocrinology, Children's Hospital at Montefiore, Bronx, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chen X, Lee HF, Zaro JL, Shen WC. Effects of receptor binding on plasma half-life of bifunctional transferrin fusion proteins. Mol Pharm 2011; 8:457-65. [PMID: 21291258 DOI: 10.1021/mp1003064] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In contrast to the wide applications of recombinant bifunctional fusion proteins in clinical usage, the systematic study for the pharmacokinetics (PK) of bifunctional fusion proteins is left blank. In this report, recombinant fusion proteins consisting of transferrin (Tf) and growth hormone (GH) or granulocyte colony-stimulating factor (G-CSF) have been constructed as a model for studying the PK of bifunctional fusion proteins. The results showed that the insertion of different linkers between the two protein domains altered the binding affinities of the fusion proteins to both domain receptors, and that the fusion proteins' plasma half-lives were greatly affected. A strong correlation between GH receptor binding affinity and plasma half-life of GH-Tf fusion proteins was observed. In addition, we demonstrated that the intracellular processing after receptor binding plays an important role in determining the half-life of fusion proteins. While the binding of the GH domain to the GH receptor will lead to endocytosis and lysosomal degradation in target cells, binding of the Tf domain to the Tf receptor may recycle the fusion protein and prolong its plasma half-life. To further confirm the effects of receptor binding on plasma half-life, G-CSF-Tf bifunctional fusion proteins with the same three linkers as GH-Tf were evaluated. While the 3 fusion proteins showed a similar G-CSF receptor binding affinity, the G-CSF-Tf fusion protein with the higher Tf receptor binding affinity exhibited longer plasma half-life. The linker insertion further demonstrated the involvement of Tf in recycling and prolonging plasma half-life. Based on our results, a model was developed to summarize the factors in determining the PK of bifunctional fusion proteins. Our findings are useful for predicting the plasma half-lives, as well as for improving the pharmacokinetic profiles of therapeutic bifunctional fusion proteins by applying linker technology.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, Los Angeles, California 90089-9121, USA
| | | | | | | |
Collapse
|
19
|
Vilardaga JP, Romero G, Friedman PA, Gardella TJ. Molecular basis of parathyroid hormone receptor signaling and trafficking: a family B GPCR paradigm. Cell Mol Life Sci 2011; 68:1-13. [PMID: 20703892 PMCID: PMC3568769 DOI: 10.1007/s00018-010-0465-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 07/06/2010] [Accepted: 07/09/2010] [Indexed: 12/14/2022]
Abstract
The parathyroid hormone (PTH) receptor type 1 (PTHR), a G protein-coupled receptor (GPCR), transmits signals to two hormone systems-PTH, endocrine and homeostatic, and PTH-related peptide (PTHrP), paracrine-to regulate different biological processes. PTHR responds to these hormonal stimuli by activating heterotrimeric G proteins, such as G(S) that stimulates cAMP production. It was thought that the PTHR, as for all other GPCRs, is only active and signals through G proteins on the cell membrane, and internalizes into a cell to be desensitized and eventually degraded or recycled. Recent studies with cultured cell and animal models reveal a new pathway that involves sustained cAMP signaling from intracellular domains. Not only do these studies challenge the paradigm that cAMP production triggered by activated GPCRs originates exclusively at the cell membrane but they also advance a comprehensive model to account for the functional differences between PTH and PTHrP acting through the same receptor.
Collapse
Affiliation(s)
- Jean-Pierre Vilardaga
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | |
Collapse
|
20
|
Ferrari SL, Bouxsein ML. Beta-arrestin-biased parathyroid hormone ligands: a new approach to the development of agents that stimulate bone formation. Sci Transl Med 2010; 1:1ps1. [PMID: 20368152 DOI: 10.1126/scitranslmed.3000268] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Because daily treatment with parathyroid hormone (PTH) increases bone mass and decreases fracture risk, physicians use this agent to treat osteoporosis. However, PTH stimulates both bone-forming and bone-resorbing cells, complicating its clinical use. New results show that, in mice, a so-called biased agonist (PTH-betaarr) that selectively activates beta-arrestin -dependent signaling leads to PTH-induced trabecular bone formation without a simultaneous increase in bone resorption. This targeted approach may pave the way for future pharmacological developments in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Serge L Ferrari
- Service of Bone Diseases, World Health Organization Collaborating Center for Osteoporosis Prevention, Department of Rehabilitation and Geriatrics, Geneva University Hospital, Geneva, Switzerland
| | | |
Collapse
|
21
|
Abstract
Postmenopausal osteoporosis (PMO) is a common skeletal disease with serious consequences due to fractures, including increased risk of disability and death. The risk of fractures can be reduced with medications that are currently available; however, these drugs are frequently not prescribed due to failure to recognize that a patient is at high risk for fracture; fear of adverse drug effects; or, sometimes, high cost. When these drugs are prescribed, long-term adherence to therapy is poor. Efforts to improve the clinical effectiveness of pharmacological therapies have included lengthening the interval between doses, simplifying drug administration, and manipulating the molecular structure of drugs in existing therapeutic classes. Recent improvement in understanding the pathophysiology of PMO at the molecular level has fostered the development of new therapeutic agents with novel mechanisms of action. This is a review of the data on the efficacy and safety of emerging drugs for the treatment of PMO, including agents with novel mechanisms of action (denosumab, odanacatib, antibody to sclerostin), new estrogen agonists/antagonists (lasofoxifene, bazedoxifene, arzoxifene), new delivery systems for existing drugs (salmon calcitonin, teriparatide), and drug combinations given concurrently, sequentially, or cyclically. These new therapeutic agents, new delivery systems, and new methods of combining drugs may ultimately reduce the great personal and economic burden of osteoporotic fractures.
Collapse
Affiliation(s)
- E Michael Lewiecki
- New Mexico Clinical Research & Osteoporosis Center, 300 Oak Street NE, Albuquerque, New Mexico 87106, USA.
| |
Collapse
|
22
|
Abstract
PTH-stimulated intracellular signaling is regulated by the cytoplasmic adaptor molecule beta-arrestin. We reported that the response of cancellous bone to intermittent PTH is reduced in beta-arrestin2(-/-) mice and suggested that beta-arrestins could influence the bone mineral balance by controlling RANKL and osteoprotegerin (OPG) gene expression. Here, we study the role of beta-arrestin2 on the in vitro development and activity of bone marrow (BM) osteoclasts (OCs) and Ephrins ligand (Efn), and receptor (Eph) mRNA levels in bone in response to PTH and the changes of bone microarchitecture in wildtype (WT) and beta-arrestin2(-/-) mice in models of bone remodeling: a low calcium diet (LoCa) and ovariectomy (OVX). The number of PTH-stimulated OCs was higher in BM cultures from beta-arrestin2(-/-) compared with WT, because of a higher RANKL/OPG mRNA and protein ratio, without directly influencing osteoclast activity. In vivo, high PTH levels induced by LoCa led to greater changes in TRACP5b levels in beta-arrestin2(-/-) compared with WT. LoCa caused a loss of BMD and bone microarchitecture, which was most prominent in beta-arrestin2(-/-). PTH downregulated Efn and Eph genes in beta-arrestin2(-/-), but not WT. After OVX, vertebral trabecular bone volume fraction and trabecular number were lower in beta-arrestin2(-/-) compared with WT. Histomorphometry showed that OC number was higher in OVX-beta-arrestin2(-/-) compared with WT. These results indicate that beta-arrestin2 inhibits osteoclastogenesis in vitro, which resulted in decreased bone resorption in vivo by regulating RANKL/OPG production and ephrins mRNAs. As such, beta-arrestins should be considered an important mechanism for the control of bone remodeling in response to PTH and estrogen deprivation.
Collapse
|
23
|
Gardella TJ. Mimetic ligands for the PTHR1: Approaches, developments, and considerations. ACTA ACUST UNITED AC 2009. [DOI: 10.1138/20090364] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
|
25
|
Prolonged signaling at the parathyroid hormone receptor by peptide ligands targeted to a specific receptor conformation. Proc Natl Acad Sci U S A 2008; 105:16525-30. [PMID: 18946036 DOI: 10.1073/pnas.0808750105] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The parathyroid hormone receptor (PTHR) is a class B G protein-coupled receptor that plays critical roles in bone and mineral ion metabolism. Ligand binding to the PTHR involves interactions to both the amino-terminal extracellular (N) domain, and transmembrane/extracellular loop, or juxtamembrane (J) regions of the receptor. Recently, we found that PTH(1-34), but not PTH-related protein, PTHrP(1-36), or M-PTH(1-14) (M = Ala/Aib(1),Aib(3),Gln(10),Har(11),Ala(12),Trp(14),Arg(19)), binds to the PTHR in a largely GTPgammaS-resistant fashion, suggesting selective binding to a novel, high-affinity conformation (R(0)), distinct from the GTPgammaS-sensitive conformation (RG). We examined the effects in vitro and in vivo of introducing the M substitutions, which enhance interaction to the J domain, into PTH analogs extended C-terminally to incorporate residues involved in the N domain interaction. As compared with PTH(1-34), M-PTH(1-28) and M-PTH(1-34) bound to R(0) with higher affinity, produced more sustained cAMP responses in cells, formed more stable complexes with the PTHR in FRET and subcellular localization assays, and induced more prolonged calcemic and phosphate responses in mice. Moreover, after 2 weeks of daily injection in mice, M-PTH(1-34) induced larger increases in trabecular bone volume and greater increases in cortical bone turnover, than did PTH(1-34). Thus, the putative R(0) PTHR conformation can form highly stable complexes with certain PTH ligand analogs and thereby mediate surprisingly prolonged signaling responses in bone and/or kidney PTH target cells. Controlling, via ligand analog design, the selectivity with which a PTH ligand binds to R(0), versus RG, may be a strategy for optimizing signaling duration time, and hence therapeutic efficacy, of PTHR agonist ligands.
Collapse
|
26
|
Martin TJ, Sims NA, Ng KW. Regulatory pathways revealing new approaches to the development of anabolic drugs for osteoporosis. Osteoporos Int 2008; 19:1125-38. [PMID: 18338097 DOI: 10.1007/s00198-008-0575-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 01/15/2008] [Indexed: 12/17/2022]
Abstract
The understanding of cell interactions and genetic controls of bone cells has provided new approaches to drug development for osteoporosis. Current emphasis in the development of new anabolic therapies is directed at modifying the effects of Wnt signalling on osteoblast differentiation and bone formation. Local signalling that results in bone formation during remodelling takes place in several ways. Growth factors released from resorbed bone matrix can contribute to preosteoblast differentiation and bone formation. Osteoclasts in the bone multicellular units (BMUs) might also generate activity that contributes to bone formation. The preosteoblasts themselves, growing in the resorption space, can communicate through cell contact and paracrine signalling mechanisms to differentiate. Osteocytes can sense the need for bone repair by detecting damage and pressure changes, and signalling to surface cells to respond appropriately. These recent insights into cell communication, together with discoveries from human and mouse genetics, have opened new pathways to drug development for osteoporosis. With the anabolic effect of parathyroid hormone on the skeleton having been established, human genetics revealed the major role of Wnt signalling in bone formation, and this has become the target of activity. Current approaches include activation at any of several points in the Wnt pathway, and neutralization of sclerostin, the protein product of the SOST gene that is produced in osteocytes as a powerful inhibitor of bone formation.
Collapse
Affiliation(s)
- T J Martin
- St Vincent's Institute of Medical Research, Melbourne, Australia.
| | | | | |
Collapse
|
27
|
|
28
|
Goltzman D. Studies on the mechanisms of the skeletal anabolic action of endogenous and exogenous parathyroid hormone. Arch Biochem Biophys 2008; 473:218-24. [PMID: 18358824 DOI: 10.1016/j.abb.2008.03.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 03/03/2008] [Accepted: 03/04/2008] [Indexed: 11/30/2022]
Abstract
Parathyroid hormone (PTH) has been viewed as catabolic for bone. Nevertheless, exogenous PTH is anabolic when administered intermittently, at a frequency that permits complete clearance between doses. In the fetus and neonate, endogenous PTH is required for normal trabecular bone formation. In older animals PTH produces net bone loss in fulfilling its calcium homeostatic role, whereas PTH-related peptide (PTHrP), acting in a paracrine/autocrine mode, is anabolic. The proliferative, differentiating, and anti-apoptotic effects of PTH on cells of the osteoblast lineage leading to anabolism can be direct, or indirect via release of local growth factors. The anabolic effect of PTH is also influenced by osteoclastic activity such that suppression of osteoclasts with anti-resorptive agents, concomitant to administering PTH, may enhance the anabolic effect by delaying a reactive osteoclastic response. In contrast, prolonged suppression of osteoclast activity prior to administering PTH appears to diminish molecular signals that increase the osteoblast pool and thereby reduces the anabolic efficacy of PTH. These observations may define the proper timing of the use of PTH as a therapeutic in diseases of bone loss. Finally, the capacity of exogenous PTH to modulate extra-osseous factors such as 1,25 dihydroxyvitamin D may also modulate its potency as an anabolic agent.
Collapse
Affiliation(s)
- David Goltzman
- Department of Medicine, McGill University and McGill University Health Centre, Montreal, Que., Canada.
| |
Collapse
|