Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Microbiome ; 10(1): 105, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35799219

ABSTRACT

BACKGROUND: Inflammatory bowel diseases (IBDs) including Crohn's disease (CD) and ulcerative colitis (UC) are characterized by chronic and debilitating gut inflammation. Altered bacterial communities of the intestine are strongly associated with IBD initiation and progression. The gut virome, which is primarily composed of bacterial viruses (bacteriophages, phages), is thought to be an important factor regulating and shaping microbial communities in the gut. While alterations in the gut virome have been observed in IBD patients, the contribution of these viruses to alterations in the bacterial community and heightened inflammatory responses associated with IBD patients remains largely unknown. RESULTS: Here, we performed in vivo microbial cross-infection experiments to follow the effects of fecal virus-like particles (VLPs) isolated from UC patients and healthy controls on bacterial diversity and severity of experimental colitis in human microbiota-associated (HMA) mice. Shotgun metagenomics confirmed that several phages were transferred to HMA mice, resulting in treatment-specific alterations in the gut virome. VLPs from healthy and UC patients also shifted gut bacterial diversity of these mice, an effect that was amplified during experimental colitis. VLPs isolated from UC patients specifically altered the relative abundance of several bacterial taxa previously implicated in IBD progression. Additionally, UC VLP administration heightened colitis severity in HMA mice, as indicated by shortened colon length and increased pro-inflammatory cytokine production. Importantly, this effect was dependent on intact VLPs. CONCLUSIONS: Our findings build on recent literature indicating that phages are dynamic regulators of bacterial communities in the gut and implicate the intestinal virome in modulating intestinal inflammation and disease. Video Abstract.


Subject(s)
Bacteriophages , Colitis, Ulcerative , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Bacteria/genetics , Bacteriophages/genetics , Colitis/therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/therapy , Inflammation , Inflammatory Bowel Diseases/microbiology , Mice
2.
MAbs ; 14(1): 1979447, 2022.
Article in English | MEDLINE | ID: mdl-34923919

ABSTRACT

Targeting immune checkpoint receptors expressed in the T cell synapse induces active and long-lasting antitumor immunity in preclinical tumor models and oncology patients. However, traditional nonhuman primate (NHP) studies in healthy animals have thus far demonstrated little to no pharmacological activity or toxicity for checkpoint inhibitors (CPIs), likely due to a quiescent immune system. We developed a NHP vaccine challenge model in Mauritius cynomolgus monkey (MCMs) that elicits a strong CD8+ T cell response to assess both pharmacology and safety within the same animal. MHC I-genotyped MCMs were immunized with three replication incompetent adenovirus serotype 5 (Adv5) encoding Gag, Nef and Pol simian immunodeficiency virus (SIV) proteins administered 4 weeks apart. Immunized animals received the anti-PD-L1 atezolizumab or an immune checkpoint-targeting bispecific antibody (mAbX) in early development. After a single immunization, Adv5-SIVs induced T-cell activation as assessed by the expression of several co-stimulatory and co-inhibitory molecules, proliferation, and antigen-specific T-cell response as measured by a Nef-dependent interferon-γ ELIspot and tetramer analysis. Administration of atezolizumab increased the number of Ki67+ CD8+ T cells, CD8+ T cells co-expressing TIM3 and LAG3 and the number of CD4+ T cells co-expressing 4-1BB, BTLA, and TIM3 two weeks after vaccination. Both atezolizumab and mAbX extended the cytolytic activity of the SIV antigen-specific CD8+ T cell up to 8 weeks. Taken together, this vaccine challenge model allowed the combined study of pharmacology and safety parameters for a new immunomodulatory protein-based therapeutic targeting CD8+ T cells in an NHP model.


Subject(s)
Adenoviridae , CD8-Positive T-Lymphocytes/immunology , SAIDS Vaccines , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/immunology , Animals , Drug Evaluation , Macaca fascicularis , Male , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , SAIDS Vaccines/pharmacology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics
4.
Regul Toxicol Pharmacol ; 81: 212-222, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27569204

ABSTRACT

Romosozumab is a humanized immunoglobulin G2 monoclonal antibody that binds and blocks the action of sclerostin, a protein secreted by the osteocyte and an extracellular inhibitor of canonical Wnt signaling. Blockade of sclerostin binding to low-density lipoprotein receptor-related proteins 5 and 6 (LRP5 and LRP6) allows Wnt ligands to activate canonical Wnt signaling in bone, increasing bone formation and decreasing bone resorption, making sclerostin an attractive target for osteoporosis therapy. Because romosozumab is a bone-forming agent and an activator of canonical Wnt signaling, questions have arisen regarding a potential carcinogenic risk. Weight-of-evidence factors used in the assessment of human carcinogenic risk of romosozumab included features of canonical Wnt signaling, expression pattern of sclerostin, phenotype of loss-of-function mutations in humans and mice, mode and mechanism of action of romosozumab, and findings from romosozumab chronic toxicity studies in rats and monkeys. Although the weight-of-evidence factors supported that romosozumab would pose a low carcinogenic risk to humans, the carcinogenic potential of romosozumab was assessed in a rat lifetime study. There were no romosozumab-related effects on tumor incidence in rats. The findings of the lifetime study and the weight-of-evidence factors collectively indicate that romosozumab administration would not pose a carcinogenic risk to humans.


Subject(s)
Antibodies, Monoclonal/toxicity , Neoplasms/chemically induced , Animals , Antibodies, Monoclonal/administration & dosage , Carcinogenicity Tests , Dose-Response Relationship, Drug , Humans , Mice , Rats , Risk Assessment
5.
J Orthop Res ; 34(1): 72-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26178170

ABSTRACT

The effects of the cathepsin K inhibitor odanacatib (ODN) on fracture healing were monitored for ~6 and 15 weeks post-fracture in two separate studies using the unilateral transverse mid-ulnar osteotomy model in skeletally mature female rabbits. Rabbits were pre-treated for 3-4 weeks with vehicle (Veh), ODN (2 mg/kg, po, daily), or alendronate (ALN) (0.3 mg/kg, sc, twice-weekly) prior to osteotomy. In Study 1, the animals were maintained on the same respective treatment for ~6 weeks. In Study 2, the animals were also continued on the same therapy or switched from Veh to ODN or ODN to Veh for 15 weeks. No treatment-related impairment of fracture union was seen by qualitative histological assessments in the first study. Cartilage retention was detected in the calluses of ALN-treated rabbits at week-6, while calluses in the ODN and Veh groups contained bony tissue with significantly less residual cartilage. ODN treatment also markedly increased the number of cathepsin K-(+) osteoclasts in the callus, indicating enhanced callus remodeling. From the second study, ex vivo DXA and pQCT confirmed that ODN treatment pre- and post-osteotomy increased callus bone mineral content and bone mineral density (BMD) versus Veh (p < 0.001) and discontinuation of ODN post-surgery returned callus BMD to Veh. Peak load of ODN- or ALN-treated calluses were comparable to Veh. ODN increased callus yield load (20%, p = 0.056) and stiffness (26%, p < 0.05) versus Veh. These studies demonstrated that ODN increased mineralized callus during the early phase of fracture repair without impairing callus formation or biomechanical integrity at the fracture site.


Subject(s)
Biphenyl Compounds/therapeutic use , Bony Callus/drug effects , Calcification, Physiologic/drug effects , Fracture Healing/drug effects , Alendronate/pharmacology , Alendronate/therapeutic use , Animals , Biphenyl Compounds/pharmacology , Female , Osteotomy , Rabbits , Random Allocation , Ulna
6.
J Bone Miner Metab ; 33(6): 625-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25534548

ABSTRACT

Pioglitazone, the peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonist is an effective therapy for type 2 diabetes, but has been associated with increased risk for bone fracture. Preclinical studies suggest that PPAR-α agonists (e.g., fenofibrate) increase bone mineral density/content, although clinical data on bone effects of fibrates are lacking. We investigated the effects of pioglitazone (10 mg/kg/day) and fenofibrate (25 mg/kg/day) on bone strength and bone histomorphometric parameters in osteopenic ovariectomized (OVX) rats. An additional group of rats received a combination of pioglitazone + fenofibrate to mimic the effects of a dual PPAR-α/γ agonist. The study consisted of a 13-week treatment phase followed by a 6-week treatment-free recovery period. Pioglitazone significantly reduced biomechanical strength at the lumbar spine and femoral neck compared with rats administered fenofibrate. Co-treatment with pioglitazone + fenofibrate had no significant effect on bone strength in comparison with OVX vehicle controls. Histomorphometric analysis of the proximal tibia revealed that pioglitazone suppressed bone formation and increased bone resorption at both cancellous and cortical bone sites relative to OVX vehicle controls. In contrast, fenofibrate did not affect bone resorption and only slightly suppressed bone formation. Discontinuation of pioglitazone treatment, both in the monotherapy and in the combination therapy arms, resulted in restoration of bone formation and resorption rates, demonstrating reversibility of effects. The above data support the concept that dual activation of PPAR-γ and PPAR-α attenuates the negative effects of PPAR-γ agonism on bone strength.


Subject(s)
Bone and Bones/pathology , Bone and Bones/physiopathology , Fenofibrate/administration & dosage , Fenofibrate/pharmacology , Ovariectomy , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Absorptiometry, Photon , Animals , Biomechanical Phenomena/drug effects , Compressive Strength/drug effects , Densitometry , Diaphyses/diagnostic imaging , Diaphyses/drug effects , Diaphyses/pathology , Diaphyses/physiopathology , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Femur/physiopathology , Femur Neck/diagnostic imaging , Femur Neck/drug effects , Femur Neck/pathology , Femur Neck/physiopathology , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Pioglitazone , Rats, Sprague-Dawley , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/pathology , Tibia/physiopathology , Tomography, X-Ray Computed
7.
J Bone Miner Res ; 30(4): 657-69, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25369992

ABSTRACT

Postmenopausal osteoporosis is a chronic disease wherein increased bone remodeling reduces bone mass and bone strength. Antiresorptive agents including bisphosphonates are commonly used to mitigate bone loss and fracture risk. Osteoclast inhibition via denosumab (DMAb), a RANKL inhibitor, is a newer approach for reducing fracture risk in patients at increased risk for fracture. The safety of transitioning from bisphosphonate therapy (alendronate; ALN) to DMAb was examined in mature ovariectomized (OVX) cynomolgus monkeys (cynos). One day after OVX, cynos (7-10/group) were treated with vehicle (VEH, s.c.), ALN (50 µg/kg, i.v., twice monthly) or DMAb (25 mg/kg/month, s.c.) for 12 months. Other animals received VEH or ALN for 6 months and then transitioned to 6 months of DMAb. DMAb caused significantly greater reductions in serum CTx than ALN, and transition from ALN to DMAb caused further reductions relative to continued ALN. DMAb and ALN decreased serum calcium (Ca), and transition from ALN to DMAb resulted in a lesser decline in Ca relative to DMAb or to VEH-DMAb transition. Bone histomorphometry indicated significantly reduced trabecular and cortical remodeling with DMAb or ALN. Compared with ALN, DMAb caused greater reductions in osteoclast surface, eroded surface, cortical porosity and fluorochrome labeling, and transition from ALN to DMAb reduced these parameters relative to continued ALN. Bone mineral density increased in all active treatment groups relative to VEH controls. Destructive biomechanical testing revealed significantly greater vertebral strength in all three groups receiving DMAb, including those receiving DMAb after ALN, relative to VEH controls. Bone mass and strength remained highly correlated in all groups at all tested skeletal sites, consistent with normal bone quality. These data indicate that cynos transitioned from ALN to DMAb exhibited reduced bone resorption and cortical porosity, and increased BMD and bone strength, without deleterious effects on Ca homeostasis or bone quality.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Bone and Bones/drug effects , Calcium/metabolism , Denosumab/pharmacology , Homeostasis/drug effects , Ovariectomy , Absorptiometry, Photon , Alendronate/adverse effects , Animals , Bone Density Conservation Agents/adverse effects , Bone and Bones/physiology , Denosumab/adverse effects , Female , Macaca fascicularis
8.
Calcif Tissue Int ; 96(1): 45-55, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25467010

ABSTRACT

The purpose of this study is to estimate the efficacy of eldecalcitol (1α, 25-Dihydroxy-2ß- (3-hydroxypropyloxy) vitamin D3; ELD) on bone metabolism after long-term administration. Six-month-old Wistar-Imamichi rats were ovariectomized (OVX) and administered ELD orally at doses of 7.5, 15, or 30 ng/kg daily. Bone mineral density (BMD), urinary excretion of deoxypyridinoline (DPD), a bone resorption marker, and serum total alkaline phosphatase (ALP), a surrogate marker of bone formation, were assessed after 3, 6, and 12 months of treatment. After 12 months of treatment, the biomechanical strength of the L4 lumbar vertebra and femoral shaft was measured, and bone histomorphometry was performed on the L3 lumbar vertebra and the tibia diaphysis. ELD prevented OVX-induced decreases in BMD of the lumbar vertebrae and femur throughout the treatment period. ELD significantly suppressed OVX-induced increases in urinary DPD excretion throughout the treatment period with minimal effects on ALP. OVX resulted in significant decreases in ultimate load and stiffness of the L4 lumbar vertebra and femoral shaft, and ELD significantly prevented the reduction in these biomechanical parameters. Bone histomorphometry at the L3 lumbar vertebra revealed that OVX induced increases in bone resorption parameters (osteoclast surface and osteoclast number) and bone formation parameters (osteoblast surface, osteoid surface, and bone formation rate), and ELD suppressed these parameters after 12 months treatment. Activation frequency, which was elevated in the OVX/vehicle group, was significantly suppressed to baseline levels in ELD-treated groups, indicating that ELD maintained bone turnover at a normal level. ELD also prevented OVX-induced deterioration of microstructure in trabecular and cortical bone. These results indicated that long-term treatment of OVX rats with ELD suppressed bone turnover, and prevented OVX-induced bone loss, deterioration of bone microstructure, and reduction in bone biomechanical strength.


Subject(s)
Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Bone and Bones/drug effects , Osteogenesis/drug effects , Vitamin D/analogs & derivatives , Animals , Bone Density/physiology , Bone Resorption/drug therapy , Female , Femur/drug effects , Lumbar Vertebrae/drug effects , Ovariectomy/methods , Rats , Time , Vitamin D/administration & dosage , Vitamin D/therapeutic use
9.
Bone ; 66: 72-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24928497

ABSTRACT

Cathepsin K (CatK) is a cysteine protease, expressed predominantly in osteoclasts (OC) which degrades demineralized bone matrix. Novel selective inhibitors of CatK are currently being developed for the treatment of postmenopausal osteoporosis. Pharmacological inhibition of CatK reduces OC resorption activity while preserving bone formation in preclinical models. Disruption of the CatK gene in mice also results in high bone mass due to impaired bone resorption and elevated formation. Here, we assessed mid-shaft femoral fracture healing in 8-10week old CatK knock-out (KO) versus wild type (WT) mice. Fracture healing and callus formation were determined in vivo weekly via X-ray, and ex vivo at days 14, 18, 28 and 42 post-fracture by radiographic scoring, micro-computed tomography (µCT), histomorphometry and terminal mechanical four point bend strength testing. Radiological evaluation indicated accelerated bone healing and remodeling for CatK KO animals based on increased total radiographic scores that included callus opacity and bridging at days 28 and 42 post-fracture. Micro-CT based total callus volume was similar in CatK KO and WT mice at day 14. Callus size in CatK KO mice was 25% smaller than that in WT mice at day 18, statistically significant by day 28 and exhibited significantly higher mineralized tissue volume and volumetric BMD as compared to WT by day 18 onward. Osteoclast surface and osteoid surface trended higher in CatK KO calluses at all time-points and osteoblast number was also significantly increased at day 28. Increased CatK KO callus mineral density was reflected in significant increases in peak load and stiffness over WT at day 42 post-fracture. Regression analysis indicated a positive correlation (r=0.8671; p<0.001) between callus BMC and peak load indicating normal mineral properties in CatK KO calluses. Taken together, gene deletion of cathepsin K in mice accelerated callus size resolution, significantly increased callus mineralized mass, and improved mechanical strength as compared to wild type mice.


Subject(s)
Bony Callus/pathology , Bony Callus/physiopathology , Calcification, Physiologic , Cathepsin K/deficiency , Femoral Fractures/pathology , Femoral Fractures/physiopathology , Animals , Biomechanical Phenomena , Bone Remodeling , Cathepsin K/metabolism , Cell Count , Female , Femoral Fractures/diagnostic imaging , Fracture Healing , Gene Deletion , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Osteoblasts/pathology , Osteoclasts/pathology , Radiography
10.
J Bone Miner Res ; 29(8): 1847-58, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24591096

ABSTRACT

Treatment with the cathepsin K (CatK) inhibitor odanacatib (ODN) protects against bone loss and maintains normal biomechanical properties in the spine and hip of ovariectomized (OVX) preclinical models. Here, we characterized the effects of ODN on the dynamics of cortical modeling and remodeling, and dimension and strength of the central femur in adult OVX-rhesus monkeys. Animals were treated with vehicle or ODN (6 or 30 mg/kg, once per day [q.d., p.o.]) in prevention mode for 21 months. Calcein and tetracycline double-labeling were given at 12 and 21 months, and the femoral cross-sections were subjected to dynamic histomorphometric and cement line analyses. ODN treatment significantly increased periosteal and endocortical bone formation (BFR/BS), accompanied with an increase in endocortical mineralizing surface (102%, p < 0.01) with the 6 mg/kg dose. ODN at both doses reduced remodeling hemiosteon numbers by 51% and 66% (p < 0.05), respectively, and ODN 30 mg/kg numerically reduced activation frequency without affecting wall thickness. On the same endocortical surface, ODN increased all modeling-based parameters, while reducing intracortical remodeling, consistent with the observed no treatment effects on cortical porosity. ODN 30 mg/kg markedly increased cortical thickness (CtTh, p < 0.001) and reduced marrow area (p < 0.01). Lastly, ODN treatment increased femoral structural strength (p < 0.001). Peak load was positively correlated with the increases in bone mineral content (BMC) (r(2) = 0.9057, p < 0.0001) and CtTh (r2 = 0.6866, p < 0.0001). Taken together, by reducing cortical remodeling-based and stimulating modeling-based bone formation, ODN significantly improved cortical dimension and strength in OVX monkeys. This novel mechanism of CatK inhibition in stimulating cortical formation suggests that ODN represents a novel therapeutic approach for the treatment of osteoporosis.


Subject(s)
Biphenyl Compounds/pharmacology , Bone Density/drug effects , Bone Remodeling/drug effects , Cathepsin K/antagonists & inhibitors , Osteogenesis/drug effects , Animals , Biphenyl Compounds/administration & dosage , Female , Hip/pathology , Macaca mulatta , Ovariectomy , Spine/drug effects
11.
Birth Defects Res B Dev Reprod Toxicol ; 98(5): 374-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24323897

ABSTRACT

BACKGROUND: The timing and duration of letrozole administration was designed to encompass the majority of postnatal development in the rat with the intent of evaluating the potential for a broad range of effects but with emphasis on expected effects on skeletal maturation. METHODS: Sprague-Dawley rats were administered letrozole via oral gavage at doses of 0.003, 0.03, and 0.3 mg/kg/day beginning on postpartum day (PPD) 7 through 91 followed by a 6-week recovery period. Clinical signs, body weight, food consumption, developmental endpoints, bone, ophthalmology, behavioral assessments, clinical/anatomic pathology, toxicokinetics, and reproductive assessments were conducted. RESULTS: Growth (body weight gain and crown-to-rump length) and food consumption were increased in females at ≥0.03 mg/kg/day and decreased in males at ≥0.003 mg/kg/day. Delayed sexual maturation in both sexes and adverse effects on reproductive function occurred at all doses. Effects on bone growth and maturation were noted in both sexes at all doses. Evidence of recovery was noted for males at 0.003 mg/kg/day and females at 0.003 and 0.03 mg/kg/day upon withdrawal of treatment. Histopathological changes in the pituitary-adrenal-gonadal axis correlated with effects on reproductive function. CONCLUSIONS: The observed effects in juvenile rats were considered predictable and primarily related to the mechanism of action of letrozole upon estrogen synthesis.


Subject(s)
Aromatase Inhibitors/toxicity , Nitriles/toxicity , Triazoles/toxicity , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Bone Development/drug effects , Dose-Response Relationship, Drug , Eating/drug effects , Female , Letrozole , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Spermatozoa/drug effects
12.
J Bone Miner Res ; 28(7): 1653-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23456892

ABSTRACT

Rosiglitazone (RSG) is an antidiabetic drug that has been associated with increased peripheral fractures, primarily in postmenopausal women. In this report, we investigated the underlying mechanisms of RSG-associated bone loss in ovariectomized (OVX) rats and determined whether changes in bone parameters associated with RSG administration are reversible on treatment cessation or preventable by coadministration with an antiresorptive agent. Nine-month-old Sprague-Dawley rats underwent OVX or sham operation. Sham-operated rats received oral vehicle only; OVX animals were randomized to receive vehicle, RSG, alendronate (ALN), or RSG plus ALN for 12 weeks. All treatment started the day after ovariectomy. After the 12-week treatment period, the OVX and RSG groups also underwent an 8-week treatment-free recovery period. Bone densitometry measurements, bone turnover markers, biomechanical testing, and histomorphometric analysis were conducted. Microcomputed tomography was also used to investigate changes in microarchitecture. RSG significantly increased deoxypyridinoline levels compared with OVX. Significant exacerbation of OVX-induced loss of bone mass, strength, and microarchitectural deterioration was observed in RSG-treated OVX animals compared with OVX controls. These effects were observed predominantly at sites rich in trabecular bone, with less pronounced effects in cortical bone. Coadministration of RSG and ALN prevented the bone loss associated with RSG treatment. Following cessation of RSG treatment, effects on bone mass and strength showed evidence of reversal. Thus, treatment of OVX rats with RSG results in loss of bone mass and strength, primarily at sites rich in trabecular bone, mainly due to increased bone resorption. These effects can be prevented by concomitant treatment with ALN and may be reversed following discontinuation of RSG.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Density/drug effects , Hypoglycemic Agents/pharmacology , Osteoporosis, Postmenopausal/drug therapy , Thiazolidinediones/pharmacology , Animals , Female , Fractures, Bone/etiology , Fractures, Bone/metabolism , Humans , Osteoporosis, Postmenopausal/complications , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Rats , Rats, Sprague-Dawley , Rosiglitazone , Time Factors , X-Ray Microtomography
13.
J Endocrinol ; 212(2): 179-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22062085

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) γ agonists, such as pioglitazone (Pio), improve glycemia and lipid profile but are associated with bone loss and fracture risk. Data regarding bone effects of PPARα agonists (including fenofibrate (Feno)) are limited, although animal studies suggest that Feno may increase bone mass. This study investigated the effects of a 13-week oral combination treatment with Pio (10 mg/kg per day)+Feno (25 mg/kg per day) on body composition and bone mass parameters compared with Pio or Feno alone in adult ovariectomized (OVX) rats, with a 4-week bone depletion period, followed by a 6-week treatment-free period. Treatment of OVX rats with Pio+Feno resulted in ∼50% lower fat mass gain compared with Pio treatment alone. Combination treatment with Pio+Feno partially prevented Pio-induced loss of bone mineral content (∼45%) and bone mineral density (BMD; ∼60%) at the lumbar spine. Similar effects of treatments were observed at the femur, most notably at sites rich in trabecular bone. At the proximal tibial metaphysis, concomitant treatment with Pio+Feno prevented Pio exacerbation of ovariectomy-induced loss of trabecular bone, resulting in BMD values in the Pio+Feno group comparable to OVX controls. Discontinuation of Pio or Feno treatment of OVX rats was associated with partial reversal of effects on bone loss or bone mass gain, respectively, while values in the Pio+Feno group remained comparable to OVX controls. These data suggest that concurrent/dual agonism of PPARγ and PPARα may reduce the negative effects of PPARγ agonism on bone mass.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Fenofibrate/therapeutic use , Osteoporosis, Postmenopausal/drug therapy , PPAR alpha/agonists , PPAR gamma/agonists , Thiazolidinediones/adverse effects , Adiposity/drug effects , Animals , Biomarkers/blood , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Resorption/chemically induced , Bone Resorption/etiology , Bone and Bones/chemistry , Bone and Bones/drug effects , Collagen Type I/blood , Drug Therapy, Combination , Female , Fenofibrate/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/therapeutic use , Osteocalcin/blood , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/physiopathology , Ovariectomy , Peptides/blood , Pioglitazone , Random Allocation , Rats , Thiazolidinediones/administration & dosage , Thiazolidinediones/therapeutic use
14.
Bone ; 49(3): 376-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21658483

ABSTRACT

A novel approach to menopausal therapy is the tissue selective estrogen complex (TSEC) that partners bazedoxifene (BZA) with conjugated estrogens (CE). We examined the effects of daily treatment with BZA 0.3mg/kg, CE 2.5mg/kg, or combined BZA/CE (BZA 0.1, 0.3, or 1.0mg/kg with CE 2.5mg/kg) over 12months on bone mass, bone architecture and strength, and biochemical markers of bone turnover in ovariectomized (OVX) female Sprague-Dawley rats vs OVX control rats. Total cholesterol and uterine weights were also evaluated. All BZA/CE dose combinations prevented ovariectomy-induced increases in bone turnover and significantly increased bone mineral density (BMD) at the lumbar spine, proximal femur, and tibia compared with OVX controls. All BZA/CE doses evaluated also prevented many of the ovariectomy-induced changes of the static and dynamic parameters of the cortical compartment of the tibia and the cancellous compartment of the L1 and L2 vertebrae. All BZA/CE doses increased biomechanical strength at the lumbar spine (L4) compared with OVX animals. The co-administration of BZA 0.3 and 1.0mg/kg/day with CE 2.5mg/kg/day showed a dose-dependent reduction in uterine wet weight compared with administration of CE alone. All BZA/CE doses significantly lowered total cholesterol levels compared with OVX controls. In conclusion, 12months of treatment with BZA/CE in OVX rats effectively maintained BMD, bone microstructure, and bone quality; and the pairing of BZA with CE prevented CE-induced uterine stimulation.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Estrogens, Conjugated (USP)/pharmacology , Estrogens/pharmacology , Indoles/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Biomarkers/metabolism , Bone Density/drug effects , Bone and Bones/anatomy & histology , Female , Femur/anatomy & histology , Femur/drug effects , Femur/physiology , Humans , Lumbar Vertebrae/anatomy & histology , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/physiology , Organ Size , Ovariectomy , Rats , Rats, Sprague-Dawley , Tibia/anatomy & histology , Tibia/drug effects , Tibia/physiology , Uterus/anatomy & histology , Uterus/drug effects
15.
J Bone Miner Res ; 26(5): 1012-21, 2011 May.
Article in English | MEDLINE | ID: mdl-21542004

ABSTRACT

Therapeutic enhancement of fracture healing would help to prevent the occurrence of orthopedic complications such as nonunion and revision surgery. Sclerostin is a negative regulator of bone formation, and treatment with a sclerostin monoclonal antibody (Scl-Ab) results in increased bone formation and bone mass in animal models. Our objective was to investigate the effects of systemic administration of Scl-Ab in two models of fracture healing. In both a closed femoral fracture model in rats and a fibular osteotomy model in cynomolgus monkeys, Scl-Ab significantly increased bone mass and bone strength at the site of fracture. After 10 weeks of healing in nonhuman primates, the fractures in the Scl-Ab group had less callus cartilage and smaller fracture gaps containing more bone and less fibrovascular tissue. These improvements at the fracture site corresponded with improvements in bone formation, bone mass, and bone strength at nonfractured cortical and trabecular sites in both studies. Thus the potent anabolic activity of Scl-Ab throughout the skeleton also was associated with an anabolic effect at the site of fracture. These results support the potential for systemic Scl-Ab administration to enhance fracture healing in patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Bone Density/drug effects , Femoral Fractures/physiopathology , Fracture Healing/drug effects , Glycoproteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Biomechanical Phenomena/drug effects , Diaphyses/drug effects , Diaphyses/pathology , Diaphyses/physiopathology , Disease Models, Animal , Femur/drug effects , Femur/pathology , Femur/physiopathology , Fibula/drug effects , Fibula/pathology , Fibula/physiopathology , Glycoproteins/immunology , Intercellular Signaling Peptides and Proteins , Macaca fascicularis , Male , Organ Size/drug effects , Osteogenesis/drug effects , Osteotomy , Rats , Rats, Sprague-Dawley
16.
J Bone Miner Res ; 26(2): 252-62, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20734451

ABSTRACT

Two cathepsin K inhibitors (CatKIs) were compared with alendronate (ALN) for their effects on bone resorption and formation in ovariectomized (OVX) rabbits. The OVX model was validated by demonstrating significant loss (9.8% to 12.8%) in lumbar vertebral bone mineral density (LV BMD) in rabbits at 13-weeks after surgery, which was prevented by estrogen or ALN. A potent CatKI, L-006235 (L-235), dosed at 10 mg/kg per day for 27 weeks, significantly decreased LV BMD loss (p < .01) versus OVX-vehicle control. ALN reduced spine cancellous mineralizing surface by 70%, whereas L-235 had no effect. Similarly, endocortical bone-formation rate and the number of double-labeled Haversian canals in the femoral diaphysis were not affected by L-235. To confirm the sparing effects of CatKI on bone formation, odanacatib (ODN) was dosed in food to achieve steady-state exposures of 4 or 9 µM/day in OVX rabbits for 27 weeks. ODN at both doses prevented LV BMD loss (p < .05 and p < .001, respectively) versus OVX-vehicle control to levels comparable with sham or ALN. ODN also dose-dependently increased BMD at the proximal femur, femoral neck, and trochanter. Similar to L-235, ODN did not reduce bone formation at any bone sites studied. The positive and highly correlative relationship of peak load to bone mineral content in the central femur and spine suggested that ODN treatment preserved normal biomechanical properties of relevant skeletal sites. Although CatKIs had similar efficacy to ALN in preventing bone loss in adult OVX rabbits, this novel class of antiresorptives differs from ALN by sparing bone formation, potentially via uncoupling bone formation from resorption.


Subject(s)
Bone Diseases, Metabolic/drug therapy , Bone and Bones/drug effects , Cathepsin K/antagonists & inhibitors , Alendronate/therapeutic use , Animals , Biphenyl Compounds/therapeutic use , Bone Density , Bone Resorption , Densitometry , Dose-Response Relationship, Drug , Estradiol/pharmacology , Female , Femur/drug effects , Haversian System/drug effects , Rabbits
17.
J Bone Miner Res ; 25(7): 1627-36, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20200973

ABSTRACT

We examined parathyroid and skeletal function in 3-month-old mice expressing the null mutation for 25-hydroxyvitamin D-1alpha-hydroxylase [1alpha(OH)ase(-/-)] and in mice expressing the null mutation for both the 1alpha(OH)ase and the calcium-sensing receptor [Casr(-/-)1alpha(OH)ase(-/-)] genes. On a normal diet, all mice were hypocalcemic, with markedly increased parathyroid hormone (PTH), increased trabecular bone volume, increased osteoblast activity, poorly mineralized bone, enlarged and distorted cartilaginous growth plates, and marked growth retardation, especially in the compound mutants. Osteoclast numbers were reduced in the Casr(-/-)1alpha(OH)ase(-/-) mice. On a high-lactose, high-calcium, high-phosphorus "rescue" diet, serum calcium and PTH were normal in the 1alpha(OH)ase(-/-) mice but increased in the Casr(-/-)1alpha(OH)ase(-/-) mice with reduced serum phosphorus. Growth plate architecture and mineralization were improved in both mutants, but linear growth of the double mutants remained abnormal. Mineralization of bone improved in all mice, but osteoblast activity and trabecular bone volume remained elevated in the Casr(-/-)1alpha(OH)ase(-/-) mice. These studies support a role for calcium-stimulated maturation of the cartilaginous growth plate and mineralization of the growth plate and bone and calcium-stimulated CaSR-mediated effects on bone resorption. PTH-mediated bone resorption may require calcium-stimulated CaSR-mediated enhancement of osteoclastic activity. (c) 2010 American Society for Bone and Mineral Research.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Bone Development/physiology , Calcium/metabolism , Osteoclasts/physiology , Receptors, Calcium-Sensing/physiology , Animals , Bone Density , Bone Resorption , Growth Plate/metabolism , Mice
18.
Endocrinology ; 150(11): 4835-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19819968

ABSTRACT

We examined the role of bone remodeling in the regulation of circulating concentrations of FGF23 using mouse models manifesting differing degrees of coupled and uncoupled bone turnover. Administration of the antiresorptive agent osteoprotegerin produced a profound reduction in bone resorption and formation in male and oophorectomized female mice, accompanied by an increase in serum levels of fibroblast growth factor 23 (FGF23) and a reduction in circulating 1,25-dihydroxyvitamin D [1,25(OH)(2)D]. In contrast, exogenous PTH(1-34) administration increased bone turnover and reduced circulating FGF23. In 1,25(OH)(2)D-deficient, 25-hydroxyvitamin D 1alpha-hydroxylase null mice on a high-calcium diet, endogenous PTH was elevated, bone formation but not resorption was increased, and serum FGF23 was virtually undetectable; on a rescue diet, serum calcium was normalized, PTH levels were reduced, bone formation was reduced, and serum FGF23 levels increased. After PTH treatment of wild-type mice, gene expression of dentin matrix protein 1 (DMP1) in bone was increased, whereas gene expression of FGF23 was reduced. In vitro studies in the osteoblastic cell line UMR-106 showed that externally added DMP1 could inhibit FGF23 gene expression and production stimulated by 1,25(OH)(2)D(3). The results show that osteoblastic bone formation is a potent modulator of FGF23 production and release into the circulation, suggest that the biological consequences on mineral homeostasis of circulating FGF23 may also be dependent on the prevailing rate of bone turnover, and provide evidence that DMP1 may be a direct negative regulator of FGF23 production in osteoblastic cells.


Subject(s)
Fibroblast Growth Factors/blood , Osteogenesis , Animals , Bone and Bones/metabolism , Cell Line , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Vitamin D/analogs & derivatives , Vitamin D/blood
19.
J Bone Miner Res ; 23(8): 1257-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18348699

ABSTRACT

PTH and 1,25(OH)(2)D each exert dual anabolic and catabolic skeletal effects. We assessed the potential interaction of PTH and 1,25(OH)(2)D in promoting skeletal anabolism by comparing the capacity of exogenous, intermittently injected PTH(1-34) to produce bone accrual in mice homozygous for the 1 alpha(OH)ase-null allele [1 alpha(OH)ase(-/-) mice] and in wildtype mice. In initial studies, 3-mo-old wildtype mice were either injected once daily (40 microg/kg) or infused continuously (120 microg/kg/d) with PTH(1-34) for up to 1 mo. Infused PTH reduced BMD, increased the bone resorption marker TRACP-5b, and raised serum calcium but did not increase serum 1,25(OH)(2)D. Injected PTH increased serum 1,25(OH)(2)D and BMD, raised the bone formation marker osteocalcin more than did infused PTH, and did not produce sustained hypercalcemia as did PTH infusion. In subsequent studies, 3-mo-old 1 alpha(OH)ase(-/-) mice, raised on a rescue diet, and wildtype littermates were injected with PTH(1-34) (40 microg/kg) either once daily or three times daily for 1 mo. In 1 alpha(OH)ase(-/-) mice, baseline bone volume (BV/TV) and bone formation (BFR/BS) were lower than in wildtype mice. PTH administered intermittently increased BV/TV and BFR/BS in a dose-dependent manner, but the increases were always less than in wildtype mice. These studies show that exogenous PTH administered continuously resorbs bone without raising endogenous 1,25(OH)(2)D. Intermittently administered PTH can increase bone accrual in the absence of 1,25(OH)(2)D, but 1,25(OH)(2)D complements this PTH action. An increase in endogenous 1,25(OH)(2)D may therefore facilitate an optimal skeletal anabolic response to PTH and may be relevant to the development of improved therapeutics for enhancing skeletal anabolism.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/metabolism , Parathyroid Hormone/pharmacology , Vitamin D/analogs & derivatives , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/deficiency , Animals , Biomarkers/metabolism , Bone Density/drug effects , Bone Resorption/metabolism , Calcium/blood , Densitometry , Humans , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteogenesis/drug effects , Parathyroid Hormone/administration & dosage , Phosphorus/blood , Tibia/cytology , Tibia/drug effects , Tomography, X-Ray Computed , Vitamin D/metabolism
20.
Endocrinology ; 148(6): 2613-21, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17332067

ABSTRACT

Osteoporosis is a leading public health problem. Although a major cause in women is thought to be a decline in estrogen, it has recently been proposed that FSH or follitropin is required for osteoporotic bone loss. We examined the FSH receptor null mouse (FORKO mouse) to determine whether altered ovarian function could induce bone loss independent of FSH action. By 3 months of age, FORKO mice developed age-dependent declines in bone mineral density and trabecular bone volume of the lumbar spine and femur, which could be partly reversed by ovarian transplantation. Bilateral ovariectomy reduced elevated circulating testosterone levels in FORKO mice and decreased bone mass to levels indistinguishable from those in ovariectomized wild-type controls. Androgen receptor blockade and especially aromatase inhibition each produced bone volume reductions in the FORKO mouse. The results indicate that ovarian secretory products, notably estrogen, and peripheral conversion of ovarian androgen to estrogen can alter bone homeostasis independent of any bone resorptive action of FSH.


Subject(s)
Bone and Bones/physiology , Follicle Stimulating Hormone/physiology , Homeostasis , Ovary/physiology , Androgens/pharmacology , Animals , Bone Density/genetics , Bone and Bones/drug effects , Bone and Bones/metabolism , Female , Homeostasis/genetics , Male , Mice , Mice, Knockout , Osteoporosis/pathology , Ovariectomy , Ovary/physiopathology , Ovary/transplantation , Receptors, FSH/genetics
SELECTION OF CITATIONS
SEARCH DETAIL