Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.509
Filter
1.
J Histochem Cytochem ; 72(5): 309-327, 2024 May.
Article in English | MEDLINE | ID: mdl-38725403

ABSTRACT

To clarify the cellular mechanism of cortical porosity induced by intermittent parathyroid hormone (PTH) administration, we examined the femoral cortical bone of mice that received 40 µg/kg/day (four times a day) human PTH (hPTH) (1-34). The PTH-driven cortical porosity initiated from the metaphyseal region and chronologically expanded toward the diaphysis. Alkaline phosphatase (ALP)-positive osteoblasts in the control mice covered the cortical surface, and endomucin-positive blood vessels were distant from these osteoblasts. In PTH-administered mice, endomucin-reactive blood vessels with TRAP-positive penetrated the ALP-positive osteoblast layer, invading the cortical bone. Statistically, the distance between endomucin-positive blood vessels and the cortical bone surface abated after PTH administration. Transmission electron microscopic observation demonstrated that vascular endothelial cells often pass through the flattened osteoblast layer and accompanied osteoclasts in the deep region of the cortical bone. The cell layers covering mature osteoblasts thickened with PTH administration and exhibited ALP, α-smooth muscle actin (αSMA), vascular cell adhesion molecule-1 (VCAM1), and receptor activator of NF-κB ligand (RANKL). Within these cell layers, osteoclasts were found near endomucin-reactive blood vessels. In PTH-administered femora, osteocytes secreted Dkk1, a Wnt inhibitor that affects angiogenesis, and blood vessels exhibited plasmalemma vesicle-associated protein, an angiogenic molecule. In summary, endomucin-positive blood vessels, when accompanied by osteoclasts in the ALP/αSMA/VCAM1/RANKL-reactive osteoblastic cell layers, invade the cortical bone, potentially due to the action of osteocyte-derived molecules such as DKK1.


Subject(s)
Cortical Bone , Endothelial Cells , Parathyroid Hormone , Animals , Mice , Parathyroid Hormone/pharmacology , Parathyroid Hormone/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cortical Bone/drug effects , Cortical Bone/metabolism , Porosity , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Immunohistochemistry , Femur/drug effects , Femur/blood supply , Femur/metabolism , Humans
2.
Biochem Biophys Res Commun ; 711: 149888, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38603833

ABSTRACT

OBJECTIVE: To investigate the effect of intermittent parathyroid hormone (iPTH) administration on pathological new bone formation during treatment of ankylosing spondylitis-related osteoporosis. METHODS: Animal models with pathological bone formation caused by hypothetical AS pathogenesis received treatment with iPTH. We determined the effects of iPTH on bone loss and the formation of pathological new bone with micro-computed tomography (micro-CT) and histological examination. In addition, the tamoxifen-inducible conditional knockout mice (CAGGCre-ERTM; PTHflox/flox, PTH-/-) was established to delete PTH and investigate the effect of endogenous PTH on pathological new bone formation. RESULTS: iPTH treatment significantly improved trabecular bone mass in the modified collagen-induced arthritis (m-CIA) model and unbalanced mechanical loading models. Meanwhile, iPTH treatment did not enhance pathological new bone formation in all types of animal models. Endogenous PTH deficiency had no effects on pathological new bone formation in unbalanced mechanical loading models. CONCLUSION: Experimental animal models of AS treated with iPTH show improvement in trabecular bone density, but not entheseal pathological bone formation,indicating it may be a potential treatment for inflammatory bone loss does in AS.


Subject(s)
Osteogenesis , Parathyroid Hormone , Animals , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Osteogenesis/drug effects , Mice , Osteoporosis/drug therapy , Osteoporosis/pathology , Mice, Knockout , Male , X-Ray Microtomography , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/pathology , Mice, Inbred C57BL , Disease Models, Animal , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Bone Density/drug effects
3.
Sci Adv ; 10(16): eadk8402, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640238

ABSTRACT

Osteoarthritis (OA) treatment is limited by the lack of effective nonsurgical interventions to slow disease progression. Here, we examined the contributions of the subchondral bone properties to OA development. We used parathyroid hormone (PTH) to modulate bone mass before OA initiation and alendronate (ALN) to inhibit bone remodeling during OA progression. We examined the spatiotemporal progression of joint damage by combining histopathological and transcriptomic analyses across joint tissues. The additive effect of PTH pretreatment before OA initiation and ALN treatment during OA progression most effectively attenuated load-induced OA pathology. Individually, PTH directly improved cartilage health and slowed the development of cartilage damage, whereas ALN primarily attenuated subchondral bone changes associated with OA progression. Joint damage reflected early transcriptomic changes. With both treatments, the structural changes were associated with early modulation of immunoregulation and immunoresponse pathways that may contribute to disease mechanisms. Overall, our results demonstrate the potential of subchondral bone-modifying therapies to slow the progression of OA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Parathyroid Hormone , Animals , Mice , Alendronate/pharmacology , Alendronate/therapeutic use , Bone and Bones , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Bone Remodeling/drug effects , Weight-Bearing
4.
Bone Res ; 12(1): 18, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38514644

ABSTRACT

The autonomic nervous system plays a crucial role in regulating bone metabolism, with sympathetic activation stimulating bone resorption and inhibiting bone formation. We found that fractures lead to increased sympathetic tone, enhanced osteoclast resorption, decreased osteoblast formation, and thus hastened systemic bone loss in ovariectomized (OVX) mice. However, the combined administration of parathyroid hormone (PTH) and the ß-receptor blocker propranolol dramatically promoted systemic bone formation and osteoporotic fracture healing in OVX mice. The effect of this treatment is superior to that of treatment with PTH or propranolol alone. In vitro, the sympathetic neurotransmitter norepinephrine (NE) suppressed PTH-induced osteoblast differentiation and mineralization, which was rescued by propranolol. Moreover, NE decreased the PTH-induced expression of Runx2 but enhanced the expression of Rankl and the effect of PTH-stimulated osteoblasts on osteoclastic differentiation, whereas these effects were reversed by propranolol. Furthermore, PTH increased the expression of the circadian clock gene Bmal1, which was inhibited by NE-ßAR signaling. Bmal1 knockdown blocked the rescue effect of propranolol on the NE-induced decrease in PTH-stimulated osteoblast differentiation. Taken together, these results suggest that propranolol enhances the anabolic effect of PTH in preventing systemic bone loss following osteoporotic fracture by blocking the negative effects of sympathetic signaling on PTH anabolism.


Subject(s)
Anabolic Agents , Bone Resorption , Osteoporotic Fractures , Mice , Animals , Parathyroid Hormone/pharmacology , Anabolic Agents/pharmacology , Osteoporotic Fractures/drug therapy , Propranolol/pharmacology , ARNTL Transcription Factors , Bone Resorption/drug therapy , Adrenergic beta-Antagonists/pharmacology
5.
Bone ; 181: 117040, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38316336

ABSTRACT

The intermittent administration of parathyroid hormone (PTH) exerts potent bone anabolic effects, which increase bone mineral density (BMD) and reduce fracture risk in osteoporotic patients. However, the underlying mechanisms remain unclear. Tmem119 has been proposed as a factor that is closely linked to the osteoblast phenotype, and we previously reported that PTH enhanced the expression of Tmem119 in mouse osteoblastic cells. However, roles of Tmem119 in the bone anabolic effects of PTH in vivo remain unknown. We herein investigated the roles of Tmem119 in bone anabolic effects of PTH using Tmem119-deficient mice. Tmem119 deficiency significantly reduced PTH-induced increases in trabecular bone volume and cortical BMD of femurs. Effects of Tmem119 deficiency on bone mass seemed predominant in female mice. Histomorphometric analyses with calcein labeling showed that Tmem119 deficiency significantly attenuated PTH-induced increases in the rates of bone formation and mineralization as well as numbers of osteoblasts. Moreover, Tmem119 deficiency significantly blunted PTH-induced decreases in phosphorylation of ß-catenin and increases in alkaline phosphatase activity in osteoblasts. In conclusion, the present results indicate that Tmem119 is involved in bone anabolic effects of PTH through osteoblastic bone formation partly related to canonical Wnt-ß-catenin signaling in mice.


Subject(s)
Anabolic Agents , Parathyroid Hormone , Humans , Animals , Female , Mice , Parathyroid Hormone/pharmacology , Parathyroid Hormone/metabolism , Osteogenesis , Anabolic Agents/pharmacology , Anabolic Agents/metabolism , beta Catenin/metabolism , Bone and Bones/metabolism , Osteoblasts/metabolism , Bone Density , Membrane Proteins/metabolism
6.
Bone ; 181: 117042, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38360197

ABSTRACT

This study investigated the efficacy of the two FDA-approved bone anabolic ligands of the parathyroid hormone receptor 1 (PTH1R), teriparatide or human parathyroid hormone 1-34 (PTH) and abaloparatide (ABL), to restoring skeletal health using a preclinical murine model of streptozotocin-induced T1-DM. Intermittent daily subcutaneous injections of equal molar doses (12 pmoles/g/day) of PTH (50 ng/g/day), ABL (47.5 ng/g/day), or vehicle, were administered for 28 days to 5-month-old C57Bl/6 J male mice with established T1-DM or control (C) mice. ABL was superior to PTH in increasing or restoring bone mass in control or T1-MD mice, respectively, which was associated with superior stimulation of trabecular and periosteal bone formation, upregulation of osteoclastic/osteoblastic gene expression, and increased circulating bone remodeling markers. Only ABL corrected the reduction in ultimate load, which is a measure of bone strength, induced by T1-DM, and it also increased energy to ultimate load. In addition, bones from T1-DM mice treated with PTH or ABL exhibited increased ultimate stress, a material index, compared to T1-DM mice administered with vehicle. And both PTH and ABL prevented the increased expression of the Wnt antagonist Sost/sclerostin displayed by T1-DM mice. Further, PTH and ABL increased to a similar extent the circulating bone resorption marker CTX and the bone formation marker P1NP in T1-DM after 2 weeks of treatment; however, only ABL sustained these increases after 4 weeks of treatment. We conclude that at equal molar doses, ABL is more effective than PTH in increasing bone mass and restoring the cortical and trabecular bone lost with T1-DM, due to higher and longer-lasting increases in bone remodeling.


Subject(s)
Diabetes Mellitus, Type 1 , Teriparatide , Humans , Mice , Male , Animals , Infant, Newborn , Teriparatide/pharmacology , Teriparatide/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Bone Density/physiology , Parathyroid Hormone-Related Protein/pharmacology , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use
7.
J Am Chem Soc ; 146(10): 6522-6529, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38417010

ABSTRACT

Parathyroid hormone 1 receptor (PTH1R) plays a key role in mediating calcium homeostasis and bone development, and aberrant PTH1R activity underlies several human diseases. Peptidic PTH1R antagonists and inverse agonists have therapeutic potential in treating these diseases, but their poor pharmacokinetics and pharmacodynamics undermine their in vivo efficacy. Herein, we report the use of a backbone-modification strategy to design a peptidic PTH1R inhibitor that displays prolonged activity as an antagonist of wild-type PTH1R and an inverse agonist of the constitutively active PTH1R-H223R mutant both in vitro and in vivo. This peptide may be of interest for the future development of therapeutic agents that ameliorate PTH1R malfunction.


Subject(s)
Drug Inverse Agonism , Receptor, Parathyroid Hormone, Type 1 , Humans , Peptides , Parathyroid Hormone/pharmacology
8.
ChemMedChem ; 19(5): e202300589, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38273777

ABSTRACT

We have previously shown that the small molecule hPTHR1 agonist PCO371 (1) orally and dose-dependently induces PTH-like calcemic and hypophostemic activity in thyroparathyroidectomized rats. Compound 2a, bearing a bicyclic aromatic ring, was identified as a novel hPTHR1 agonist during hit to lead modification. It showed moderate PTHR1 agonistic activity with an EC20 value of 15 µM, and its metabolic stability in human liver microsome (hLM) as well as its solubility in phosphate buffer (PPb) and Fasted state simulated intestinal fluid (FaSSIF) were found to be poor. As results of the initial derivatization of 2a, we identified the indole derivatives as another scaffold. In this article, we report on the structure-activity relationship (SAR), structure-metabolism relationship (SMR), and structure-solubility relationship (SSR) of bicyclic aromatic derivatives, and the in vivo efficacy of 2j.


Subject(s)
Antipsychotic Agents , Humans , Animals , Rats , Microsomes, Liver , Solubility , Structure-Activity Relationship , Parathyroid Hormone/pharmacology
9.
Mol Cell Endocrinol ; 583: 112159, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38228226

ABSTRACT

In hyperparathyroidism (hyperPTH), excessive amounts of PTH are secreted, interfering with calcium regulation in the body. Several drugs can control the disease's side effects, but none of them is an alternative treatment to surgery. Therefore, new drug candidates are necessary. In this study, three computationally repositioned drugs, DG 041, IMD 0354, and cucurbitacin I, are evaluated in an in vitro model of hyperPTH. First, we integrated publicly available transcriptomics datasets to propose drug candidates. Using 3D spheroids derived from a single primary hyperPTH patient, we assessed their in vitro efficacy. None of the proposed drugs affected the viability of healthy cell control (HEK293) or overactive parathyroid cells at the level of toxicity. This behavior was attributed to the non-cancerous nature of the parathyroid cells, establishing the hyperPTH disease model. Cucurbitacin I and IMD 0354 exhibited a slight inverse relationship between increased drug concentrations and cell viability, whereas DG 041 increased viability. Based on these results, further studies are needed on the mechanism of action of the repurposed drugs, including determining the effects of these drugs on cellular PTH synthesis and secretion and on the metabolic pathways that regulate PTH secretion.


Subject(s)
Acrylamides , Benzamides , Hyperparathyroidism, Primary , Parathyroid Hormone , Sulfones , Triterpenes , Humans , Parathyroid Hormone/pharmacology , Parathyroid Hormone/metabolism , Calcium , Drug Repositioning , HEK293 Cells , Hyperparathyroidism, Primary/drug therapy
10.
Bone ; 179: 116983, 2024 02.
Article in English | MEDLINE | ID: mdl-38013019

ABSTRACT

Stress fractures occur as a result of repeated mechanical stress on bone and are commonly found in the load-bearing lower extremities. Macrophages are key players in the immune system and play an important role in bone remodeling and fracture healing. However, the role of macrophages in stress fractures has not been adequately addressed. We hypothesize that macrophage infiltration into a stress fracture callus site promotes bone healing. To test this, a unilateral stress fracture induction model was employed in which the murine ulna of four-month-old, C57BL/6 J male mice was repeatedly loaded with a pre-determined force until the bone was displaced a distance below the threshold for complete fracture. Mice were treated daily with parathyroid hormone (PTH, 50 µg/kg/day) starting two days before injury and continued until 24 h before euthanasia either four or six days after injury, or treated with trabectedin (0.15 mg/kg) on the day of stress fracture and euthanized three or seven days after injury. These treatments were used due to their established effects on macrophages. While macrophages have been implicated in the anabolic effects of PTH, trabectedin, an FDA approved chemotherapeutic, compromises macrophage function and reduces bone mass. At three- and four-days post injury, callus macrophage numbers were analyzed histologically. There was a significant increase in macrophages with PTH treatment compared to vehicle in the callus site. By one week of healing, treatments differentially affected the bony callus as analyzed by microcomputed tomography. PTH enhanced callus bone volume. Conversely, callus bone volume was decreased with trabectedin treatment. Interestingly, concurrent treatment with PTH and trabectedin rescued the reduction observed in the callus with trabectedin treatment alone. This study reports on the key involvement of macrophages during stress fracture healing. Given these observed outcomes on macrophage physiology and bone healing, these findings may be important for patients actively receiving either of these FDA-approved therapeutics.


Subject(s)
Fractures, Stress , Parathyroid Hormone , Humans , Male , Mice , Animals , Infant , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Trabectedin/pharmacology , Fractures, Stress/drug therapy , Fractures, Stress/pathology , X-Ray Microtomography/methods , Mice, Inbred C57BL , Bony Callus/pathology , Fracture Healing , Macrophages
11.
Arthroscopy ; 40(4): 1093-1104.e2, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38000485

ABSTRACT

PURPOSE: To investigate the effect of recombinant human parathyroid hormone (rhPTH) biocomposite on bone-to-tendon interface (BTI) healing for surgical repair of a chronic rotator cuff tear (RCT) model of rabbit, focusing on genetic, histologic, biomechanical and micro-computed tomography (CT) evaluations. METHODS: Sixty-four rabbits were equally assigned to the 4 groups: saline injection (group A), nanofiber sheet alone (group B), rhPTH-soaked nanofiber sheet (nanofiber sheet was soaked with rhPTH, group C), and rhPTH biocomposite (rhPTH permeated the nanofiber sheet by coaxial electrospinning, group D). The release kinetics of rhPTH (groups C and D) was examined for 6 weeks in vitro. Nanofiber scaffolds were implanted on the surface of the repair site 6 weeks after the induction of chronic RCT. Genetic and histologic analyses were conducted 4 weeks after surgery. Furthermore, genetic, histologic, biomechanical, micro-CT, and serologic analyses were performed 12 weeks after surgery. RESULTS: In vivo, group D showed the highest collagen type I alpha 1 (COL1A1), collagen type III alpha 1 (COL3A1), and bone morphogenetic protein 2 (BMP-2) messenger RNA (mRNA) expression levels (all P < .001) 4 weeks after surgery; however, there were no differences between groups at 12 weeks postsurgery. After 12 weeks postsurgery, group D showed better collagen fiber continuity and orientation, denser collagen fibers, more mature bone-to-tendon junction, and greater fibrocartilage layer formation compared with the other groups (all P < .05). Furthermore, group D showed the highest load-to-failure rate (28.9 ± 2.0 N/kg for group A, 30.1 ± 3.3 N/kg for group B, 39.7 ± 2.7 N/kg for group C, and 48.2 ± 4.5 N/kg for group D, P < .001) and micro-CT outcomes, including bone and tissue mineral density, and bone volume/total volume rate (all P < .001) at 12 weeks postsurgery. CONCLUSIONS: In comparison to rhPTH-soaked nanofiber sheet and the other control groups, rhPTH biocomposite effectively accelerated BTI healing by enhancing the mRNA expression levels of COL1A1, COL3A1, and BMP-2 at an early stage and achieving tenogenesis, chondrogenesis, and osteogenesis at 12 weeks after surgical repair of a chronic RCT model of rabbit. CLINICAL RELEVANCE: The present study might be a transitional study to demonstrate the efficacy of rhPTH biocomposites on BTI healing for surgical repair of chronic RCTs as an adaptable polymer biomaterial in humans.


Subject(s)
Rotator Cuff Injuries , Animals , Humans , Rabbits , Rotator Cuff Injuries/surgery , Rotator Cuff Injuries/pathology , Osteogenesis , Chondrogenesis , Wound Healing , Disease Models, Animal , Tendons/surgery , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Collagen/pharmacology , RNA, Messenger , Biomechanical Phenomena
12.
J Ultrasound Med ; 43(2): 385-395, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37994205

ABSTRACT

The intermittent injection of teriparatide, a recombinant fragment of human parathyroid hormone (PTH 1-34), activates anabolic activity on bone turnover. However, the PTH administration period is limited to 2 years. Thus, sequential therapy after discontinuation of PTH is required. Low-intensity pulsed ultrasound (LIPUS) has been widely used for bone fracture healing. In this study, we examined the effects of LIPUS on bone mass after PTH withdrawal in ovariectomized (OVX) model mice. The LIPUS-non-irradiated femoral trabecular bone mineral density (BMD) in the treated after PTH withdrawal was significantly decreased. Meanwhile, the femoral BMD in the OVX + PTH-LIPUS group was remarkably higher than that of the OVX group. Additionally, mRNA expression of Runx2, Osterix, Col1a1, and ALP increased significantly following LIPUS irradiation after PTH withdrawal. These results suggest that LIPUS protected against femoral trabecular BMD loss and up-regulated the osteogenic factors following PTH withdrawal in OVX mice.


Subject(s)
Bone Density , Parathyroid Hormone , Humans , Mice , Animals , Female , Parathyroid Hormone/pharmacology , Osteogenesis , Ultrasonic Waves , Ovariectomy
13.
Clin Oral Implants Res ; 35(3): 305-320, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38124678

ABSTRACT

OBJECTIVES: Intermittent administration of parathyroid hormone (PTH) increases systemic bone mass. However, the effect of PTH on osseous and soft tissue healing around implants in osteoporosis patients remains unclear. This study aimed to investigate the effects of PTH on tissue healing around implants in ovariectomized rats and to compare systemic and intraoral administration routes. MATERIAL AND METHODS: Implants were placed at the healed sites of ovariectomized rats 3 weeks after maxillary first molar extraction. Rats were randomly divided into two groups that received either daily systemic subcutaneous or local intraoral PTH administration. Maxillae were dissected to examine bone architectures with micro-computed tomography images. Histomorphometric and immunohistochemical analyses were performed to evaluate osseous and soft tissue healing around the implants. RESULTS: Regardless of the administration route, PTH significantly increased bone area and the numbers of osteoblasts, osteoclasts, and osteocytes in the first and second inside and outside areas of implant threads, in addition to decreasing the number of sclerostin+ osteocytes. However, the intraoral PTH administration route was superior to the systemic route by significantly improving bone quality and promoting collagen production in the connective tissue around implants. CONCLUSIONS: Parathyroid hormone administration promoted both osseous and soft tissue healing around implants, irrespective of administration route. Interestingly, intraoral administration improved the evaluated parameters more than systemic administration. Thus, the intraoral route could become a useful treatment strategy for implant treatment in osteoporosis patients.


Subject(s)
Dental Implants , Osteoporosis , Humans , Rats , Animals , Parathyroid Hormone/pharmacology , Maxilla/diagnostic imaging , Maxilla/surgery , X-Ray Microtomography
14.
Bone ; 180: 116994, 2024 03.
Article in English | MEDLINE | ID: mdl-38135023

ABSTRACT

In this study, we aimed to quantify the localised effects of mechanical loading (ML), low (20 µg/kg/day), moderate (40 µg/kg/day) or high (80 µg/kg/day) dosages of parathyroid hormone (PTH), and combined (PTHML) treatments on cortical bone adaptation in healthy 19-week old female C57BL/6 mice. To this end, we utilise a previously reported image analysis algorithm on µCT data of the mouse tibia published by Sugiyama et al. (2008) to measure changes in cortical area, marrow cavity area and local cortical thickness measures (ΔCt.Ar, ΔMa.Ar, ΔCt.Th respectively), evaluated at two cross-sections within the mouse tibia (proximal-middle (37 %) and middle (50 %)), and are compared to a superposed summation (P + M) of individual treatments to determine the effectiveness of combining treatments in vivo. ΔCt.Ar analysis revealed a non-linear, synergistic interactions between PTH and ML in the 37 % cross-section that saturates at higher PTH dosages, whereas the 50 % cross-section experiences an approximately linear, additive adaptation response. This coincided with an increase in ΔMa.Ar (indicating resorption of the endosteal surface), which was only counteracted by combined high dose PTH with ML in the middle cross-section. Regional analysis of ΔCt.Th changes reveal localised cortical thinning in response to low dose PTH treatment in the posteromedial region of the middle cross-section, signifying that PTH does not provide a homogeneous adaptation response around the cortical perimeter. We observe a synergistic response in the proximal-middle cross-section, with regions of compressive strain experiencing the greatest adaptation response to PTHML treatments, (peak ΔCt.Th of 189.32, 213.78 and 239.30 µm for low, moderate and high PTHML groups respectively). In contrast, PTHML treatments in the middle cross-section show a similar response to the superposed P + M group, with the exception of the combined high dose PTHML treatment which shows a synergistic interaction. These analyses suggest that, in mice, adding mechanical loading to PTH treatments leads to region specific bone responses; synergism of PTHML is only achieved in some regions experiencing high loading, while other regions respond additively to this combined treatment.


Subject(s)
Parathyroid Hormone , Tibia , Mice , Female , Animals , Parathyroid Hormone/pharmacology , Tibia/physiology , Mice, Inbred C57BL , Bone and Bones , Cortical Bone/diagnostic imaging , Disease Models, Animal
15.
J Orthop Res ; 42(6): 1254-1266, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38151816

ABSTRACT

Combined treatment with PTH(1-34) and mechanical loading confers increased structural benefits to bone than monotherapies. However, it remains unclear how this longitudinal adaptation affects the bone mechanics. This study quantified the individual and combined longitudinal effects of PTH(1-34) and mechanical loading on the bone stiffness and strength evaluated in vivo with validated micro-finite element (microFE) models. C57BL/6 mice were ovariectomised at 14-week-old and treated either with injections of PTH(1-34), compressive tibia loading or both interventions concurrently. Right tibiae were in vivo microCT-scanned every 2 weeks from 14 until 24-week-old. MicroCT images were rigidly registered to reference tibia and the cortical organ level (whole bone) and tissue level (midshaft) morphometric properties and bone mineral content were quantified. MicroCT images were converted into voxel-based homogeneous, linear elastic microFE models to estimate the bone stiffness and strength. This approach allowed us for the first time to quantify the longitudinal changes in mechanical properties induced by combined treatments in a model of accelerated bone resorption. Both changes of stiffness and strength were higher with co-treatment than with individual therapies, consistent with increased benefits with the tibia bone mineral content and cortical area, properties strongly associated with the tibia mechanics. The longitudinal data shows that the two bone anabolics, both individually and combined, had persistent benefit on estimated mechanical properties, and that benefits (increased stiffness and strength) remained after treatment was withdrawn.


Subject(s)
Mice, Inbred C57BL , Ovariectomy , Parathyroid Hormone , Tibia , X-Ray Microtomography , Animals , Tibia/drug effects , Tibia/diagnostic imaging , Tibia/physiology , Female , Parathyroid Hormone/pharmacology , Bone Density/drug effects , Weight-Bearing , Biomechanical Phenomena , Mice , Finite Element Analysis
16.
Sci Transl Med ; 15(722): eadg8982, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37967203

ABSTRACT

Low back pain (LBP) is one of the most prevalent diseases affecting quality of life, with no disease-modifying therapy. During aging and spinal degeneration, the balance between the normal endplate (EP) bilayers of cartilage and bone shifts to more bone. The aged/degenerated bony EP has increased porosity because of osteoclastic remodeling activity and may be a source of LBP due to aberrant sensory innervation within the pores. We used two mouse models of spinal degeneration to show that parathyroid hormone (PTH) treatment induced osteogenesis and angiogenesis and reduced the porosity of bony EPs. PTH increased the cartilaginous volume and improved the mechanical properties of EPs, which was accompanied by a reduction of the inflammatory factors cyclooxygenase-2 and prostaglandin E2. PTH treatment furthermore partially reversed the innervation of porous EPs and reversed LBP-related behaviors. Conditional knockout of PTH 1 receptors in the nucleus pulposus (NP) did not abolish the treatment effects of PTH, suggesting that the NP is not the primary source of LBP in our mouse models. Last, we showed that aged rhesus macaques with spontaneous spinal degeneration also had decreased EP porosity and sensory innervation when treated with PTH, demonstrating a similar mechanism of PTH action on EP sclerosis between mice and macaques. In summary, our results suggest that PTH treatment could partially reverse EP restructuring during spinal regeneration and support further investigation into this potentially disease-modifying treatment strategy for LBP.


Subject(s)
Low Back Pain , Parathyroid Hormone , Mice , Animals , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Macaca mulatta , Quality of Life , Disease Models, Animal
17.
J Transl Med ; 21(1): 844, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37996876

ABSTRACT

BACKGROUND: Non-union formation still represents a major burden in trauma and orthopedic surgery. Moreover, aged patients are at an increased risk for bone healing failure. Parathyroid hormone (PTH) has been shown to accelerate fracture healing in young adult animals. However, there is no information whether PTH also stimulates bone regeneration in atrophic non-unions in the aged. Therefore, the aim of the present study was to analyze the effect of PTH on bone regeneration in an atrophic non-union model in aged CD-1 mice. METHODS: After creation of a 1.8 mm segmental defect, mice femora were stabilized by pin-clip fixation. The animals were treated daily with either 200 mg/kg body weight PTH 1-34 (n = 17) or saline (control; n = 17) subcutaneously. Bone regeneration was analyzed by means of X-ray, biomechanics, micro-computed tomography (µCT) imaging as well as histological, immunohistochemical and Western blot analyses. RESULTS: In PTH-treated animals bone formation was markedly improved when compared to controls. This was associated with an increased bending stiffness as well as a higher number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and CD31-positive microvessels within the callus tissue. Furthermore, PTH-treated aged animals showed a decreased inflammatory response, characterized by a lower number of MPO-positive granulocytes and CD68-positive macrophages within the bone defects when compared to controls. Additional Western blot analyses demonstrated a significantly higher expression of cyclooxygenase (COX)-2 and phosphoinositide 3-kinase (PI3K) in PTH-treated mice. CONCLUSION: Taken together, these findings indicate that PTH is an effective pharmacological compound for the treatment of non-union formation in aged animals.


Subject(s)
Bone Regeneration , Phosphatidylinositol 3-Kinases , Humans , Mice , Animals , Aged , X-Ray Microtomography , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Fracture Healing
18.
J Mech Behav Biomed Mater ; 148: 106209, 2023 12.
Article in English | MEDLINE | ID: mdl-37918338

ABSTRACT

BACKGROUND AND OBJECTIVE: It is essential to know the quantitative interactions between biological tissues and external mechanical and chemical stimuli. This assists the physicians to better know the quantitative behavior of the tissue and plan more effective therapy. In the literature, the effect of the chemical and mechanical loading was investigated on the bone biological cell activities and some mechanical features, but a lack of prediction of bone injury under the chemical and mechanical factors was sensed. Therefore, the present study aims to investigate the effects of the application of major chemical factors involved in ossification, including RANKL1 (Receptor Activator of Nuclear Factor Kappa Beta Ligand), PTH2 (Parathyroid Hormone), and OPG3 (Osteoprotegerin) on the mandibular bone biological osteoblast and osteoclast activities and mechanical properties. Moreover, the study assesses the bone injury possibility under uniform mastication pressure applied on the premolar tooth in terms of the mechanostat theory undergoing the effects of the chemical factors. METHODS: A 3D geometry of the mandible-tooth assembly was generated from the CT image dataset. The geometry was next purified, solidified, and exported to FEM4 (Finite Element Method) software to be meshed, where boundary conditions and loading were applied. Moreover, the mathematical system of differential equations to model the chemical factor effects on osteoblast and osteoclast activities as well as bone matrix volume fraction and elastic stiffness relations were applied. Next, the values of the equivalent strain were calculated to predict the injury states of the bone. RESULTS: The results complied with the literature data. The results showed that RANKL and PTH increased the values of the equivalent strain from 450 µÎµ to 11500 µÎµ, while OPG reduced that from 450 µÎµ to 300 µÎµ. CONCLUSIONS: Therefore, RANKL and PTH doses of this study put the bone at risk of injury compared to the baseline of no dose applied, while OPG secured the bone from injury.


Subject(s)
Osteoclasts , Osteogenesis , Parathyroid Hormone/pharmacology , Osteoblasts , Mandible
19.
Front Cell Infect Microbiol ; 13: 1230568, 2023.
Article in English | MEDLINE | ID: mdl-37829606

ABSTRACT

Introduction: Diabetes mellitus (DM) impairs fracture healing and is associated with susceptibility to infection, which further inhibits fracture healing. While intermittent parathyroid hormone (1-34) (iPTH) effectively improves fracture healing, it is unknown whether infection-associated impaired fracture healing can be rescued with PTH (teriparatide). Methods: A chronic diet-induced type 2 diabetic mouse model was used to yield mice with decreased glucose tolerance and increased blood glucose levels compared to lean-fed controls. Methicillin-resistant Staphylococcus aureus (MRSA) was inoculated in a surgical tibia fracture model to simulate infected fracture, after which mice were treated with a combination of antibiotics and adjunctive teriparatide treatment. Fracture healing was assessed by Radiographic Union Scale in Tibial Fractures (RUST), micro-computed tomography (µCT), biomechanical testing, and histology. Results: RUST score was significantly poorer in diabetic mice compared to their lean nondiabetic counterparts. There were concomitant reductions in micro-computed tomography (µCT) parameters of callus architecture including bone volume/total volume, trabecular thickness, and total mineral density in type 2 diabetes mellitus (T2DM) mice. Biomechanicaltesting of fractured femora demonstrated diminished torsional rigidity, stiffness, and toughness to max torque. Adjuvant teriparatide treatment with systemic antibiotic therapy improved numerous parameters of bone microarchitecture bone volume, increased connectivity density, and increased trabecular number in both the lean and T2DM group. Despite the observation that poor fracture healing in T2DM mice was further impaired by MRSA infection, adjuvant iPTH treatment significantly improved fracture healing compared to antibiotic treatment alone in infected T2DM fractures. Discussion: Our results suggest that teriparatide may constitute a viable adjuvant therapeutic agent to improve bony union and bone microarchitecture to prevent the development of septic nonunion under diabetic conditions.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Methicillin-Resistant Staphylococcus aureus , Mice , Animals , Fracture Healing , Teriparatide/therapeutic use , Teriparatide/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , X-Ray Microtomography , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use
20.
Evol Dev ; 25(4-5): 274-288, 2023 07.
Article in English | MEDLINE | ID: mdl-37540043

ABSTRACT

Adaptation to different environments can be achieved by physiological shifts throughout development. Hormonal regulators shape the physiological and morphological traits of the evolving animals making them fit for the particular ecological surroundings. We hypothesized that the artificially induced hypersynthesis of calcitonin and parathyroid hormone mutually influencing calcium metabolism could affect bone formation during early ontogeny in fish imitating the heterochrony in craniofacial ossification in natural adaptive morphs. Conducting an experiment, we found that the long-standing treatment of salmonid juveniles with high doses of both hormones irreversibly shifts the corresponding hormone status for a period well beyond the time scale for total degradation of the injected hormone. The hormones program the ossification of the jaw suspension bones and neurocranial elements in a specific manner affecting the jaws position and pharingo-branchial area stretching. These morphological shifts resemble the adaptive variants found in sympatric pelagic and demersal morphs of salmonids. We conclude that solitary deviations in the regulators of calcium metabolism could determine functional morphological traits via transformations in skeletal development.


Subject(s)
Calcium , Salmonidae , Animals , Calcium/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone/physiology , Osteogenesis , Salmonidae/metabolism , Skull
SELECTION OF CITATIONS
SEARCH DETAIL
...