Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Sci Rep ; 11(1): 11839, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088950

ABSTRACT

Heart failure with reduced ejection fraction (HFrEF) constitutes 50% of HF hospitalizations and is characterized by high rates of mortality. To explore the underlying mechanisms of HFrEF etiology and progression, we studied the molecular and cellular differences in four chambers of non-failing (NF, n = 10) and HFrEF (n = 12) human hearts. We identified 333 genes enriched within NF heart subregions and often associated with cardiovascular disease GWAS variants. Expression analysis of HFrEF tissues revealed extensive disease-associated transcriptional and signaling alterations in left atrium (LA) and left ventricle (LV). Common left heart HFrEF pathologies included mitochondrial dysfunction, cardiac hypertrophy and fibrosis. Oxidative stress and cardiac necrosis pathways were prominent within LV, whereas TGF-beta signaling was evident within LA. Cell type composition was estimated by deconvolution and revealed that HFrEF samples had smaller percentage of cardiomyocytes within the left heart, higher representation of fibroblasts within LA and perivascular cells within the left heart relative to NF samples. We identified essential modules associated with HFrEF pathology and linked transcriptome discoveries with human genetics findings. This study contributes to a growing body of knowledge describing chamber-specific transcriptomics and revealed genes and pathways that are associated with heart failure pathophysiology, which may aid in therapeutic target discovery.


Subject(s)
Gene Expression Profiling , Heart Failure/metabolism , Heart Ventricles/metabolism , Ventricular Dysfunction, Left/metabolism , Female , Fibroblasts/metabolism , Gene Regulatory Networks , Heart Atria/physiopathology , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Hospitalization , Humans , Male , Middle Aged , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Necrosis , Oxidative Stress , Prognosis , Signal Transduction , Stroke Volume/physiology , Transcription, Genetic , Transcriptome , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/physiology
2.
Cell Rep Med ; 2(5): 100263, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095876

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) regulate glucose and energy homeostasis. Targeting both pathways with GIP receptor (GIPR) antagonist antibody (GIPR-Ab) and GLP-1 receptor (GLP-1R) agonist, by generating GIPR-Ab/GLP-1 bispecific molecules, is an approach for treating obesity and its comorbidities. In mice and monkeys, these molecules reduce body weight (BW) and improve many metabolic parameters. BW loss is greater with GIPR-Ab/GLP-1 than with GIPR-Ab or a control antibody conjugate, suggesting synergistic effects. GIPR-Ab/GLP-1 also reduces the respiratory exchange ratio in DIO mice. Simultaneous receptor binding and rapid receptor internalization by GIPR-Ab/GLP-1 amplify endosomal cAMP production in recombinant cells expressing both receptors. This may explain the efficacy of the bispecific molecules. Overall, our GIPR-Ab/GLP-1 molecules promote BW loss, and they may be used for treating obesity.


Subject(s)
Body Weight/physiology , Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test/methods , Haplorhini/metabolism , Mice, Obese
3.
PLoS One ; 15(8): e0231234, 2020.
Article in English | MEDLINE | ID: mdl-32804947

ABSTRACT

Cardiometabolic syndrome has become a global health issue. Heart failure is a common comorbidity of cardiometabolic syndrome. Successful drug development to prevent cardiometabolic syndrome and associated comorbidities requires preclinical models predictive of human conditions. To characterize the heart failure component of cardiometabolic syndrome, cardiometabolic, metabolic, and renal biomarkers were evaluated in lean and obese ZSF1 19- to 32-week-old male rats. Histopathological assessment of kidneys and hearts was performed. Cardiac function, exercise capacity, and left ventricular gene expression were also analyzed. Obese ZSF1 rats exhibited multiple features of human cardiometabolic syndrome by pathological changes in systemic renal, metabolic, and cardiovascular disease circulating biomarkers. Hemodynamic assessment, echocardiography, and decreased exercise capacity confirmed heart failure with preserved ejection fraction. RNA-seq results demonstrated changes in left ventricular gene expression associated with fatty acid and branched chain amino acid metabolism, cardiomyopathy, cardiac hypertrophy, and heart failure. Twelve weeks of growth differentiation factor 15 (GDF15) treatment significantly decreased body weight, food intake, blood glucose, and triglycerides and improved exercise capacity in obese ZSF1 males. Systemic cardiovascular injury markers were significantly lower in GDF15-treated obese ZSF1 rats. Obese ZSF1 male rats represent a preclinical model for human cardiometabolic syndrome with established heart failure with preserved ejection fraction. GDF15 treatment mediated dietary response and demonstrated a cardioprotective effect in obese ZSF1 rats.


Subject(s)
Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/pharmacology , Metabolic Syndrome/metabolism , Animals , Biomarkers/metabolism , Heart/physiology , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Kidney/metabolism , Male , Metabolic Syndrome/complications , Myocardium/metabolism , Obesity/complications , Rats , Rats, Inbred Strains , Rats, Zucker , Stroke Volume/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
4.
Regul Toxicol Pharmacol ; 115: 104697, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32590049

ABSTRACT

Romosozumab (EVENITY™ [romosozumab-aqqg in the US]) is a humanized monoclonal antibody that inhibits sclerostin and has been approved in several countries for the treatment of osteoporosis in postmenopausal women at high risk of fracture. Sclerostin is expressed in bone and aortic vascular smooth muscle (AVSM). Its function in AVSM is unclear but it has been proposed to inhibit vascular calcification, atheroprogression, and inflammation. An increased incidence of positively adjudicated serious cardiovascular adverse events driven by an increase in myocardial infarction and stroke was observed in romosozumab-treated subjects in a clinical trial comparing alendronate with romosozumab (ARCH; NCT01631214) but not in a placebo-controlled trial (FRAME; NCT01575834). To investigate the effects of sclerostin inhibition with sclerostin antibody on the cardiovascular system, a comprehensive nonclinical toxicology package with additional cardiovascular studies was conducted. Although pharmacodynamic effects were observed in the bone, there were no functional, morphological, or transcriptional effects on the cardiovascular system in animal models in the presence or absence of atherosclerosis. These nonclinical studies did not identify evidence that proves the association between sclerostin inhibition and adverse cardiovascular function, increased cardiovascular calcification, and atheroprogression.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Bone Density Conservation Agents/pharmacology , Cardiovascular System/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Bone Density Conservation Agents/therapeutic use , Drug Evaluation, Preclinical , Female , Fractures, Bone/prevention & control , Humans , Macaca fascicularis , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Osteoporosis/drug therapy , Rats, Sprague-Dawley , Risk
5.
J Bone Miner Res ; 34(1): 171-181, 2019 01.
Article in English | MEDLINE | ID: mdl-30184271

ABSTRACT

Antiresorptive agents, such as bisphosphonates and denosumab, are frequently used for the management of osteoporosis. Indeed, both medications decrease the risk of osteoporotic fractures; however, these medications are associated with rare but potentially severe side effects, such as osteonecrosis of the jaw (ONJ). ONJ, defined as an area of exposed bone in the maxillofacial region that lasts for 8 weeks, often presents with significant pain and infection and can lead to serious complications. Interestingly, other treatments for osteoporosis have been developed, such as antibodies against the osteocyte-secreted protein, sclerostin. Sclerostin functions to inhibit the Wnt signaling cascade, leading to inhibition of bone formation. In clinical trials, a sclerostin antibody (romosozumab, Amgen Inc., UCB Brussels) increases bone formation and lowers the risk of osteoporotic fractures. However, in conjunction with increased osteoblastic activity, a reduction in bone resorption markers is observed. This antiresorptive effect raises the concern of possible ONJ development in patients treated with sclerostin antibodies. Here, utilizing ligature-induced experimental periodontitis (EP), we evaluated the effects of sclerostin inhibition on the development of ONJ-like lesions in ovariectomized rats. Beginning 8 weeks post-ovariectomy, rats were treated for 22 weeks with weekly injections of vehicle (Veh), 200 µg/kg zoledronic acid (ZA), a potent bisphosphonate at 100-fold the osteoporosis dose, or 5 mg/kg sclerostin antibody (Scl-Ab) at the osteoporotic dose. EP was initiated at week 12 and maintained for the remainder of the study. Scl-Ab treatment transiently increased serum P1NP, a bone formation marker, increased BV/TV, and decreased eroded surfaces in lumbar vertebrae. ZA-treated rats developed histologic features of ONJ, whereas Veh-treated controls did not. Scl-Ab animals lost less periodontal bone in sites with EP. However, these animals presented with no histologic signs of ONJ. In conclusion, sclerostin inhibition enhanced structural bone parameters, without inducing ONJ-like lesions, in ovariectomized rats with EP. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Antibodies/pharmacology , Bisphosphonate-Associated Osteonecrosis of the Jaw/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Osteoporosis/metabolism , Periodontitis/metabolism , Animals , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Female , Genetic Markers , Osteoporosis/pathology , Ovariectomy , Periodontitis/pathology , Rats , Rats, Sprague-Dawley , Zoledronic Acid/adverse effects , Zoledronic Acid/pharmacology
6.
Bone ; 116: 162-170, 2018 11.
Article in English | MEDLINE | ID: mdl-30077758

ABSTRACT

Serum calcium (Ca) is maintained in a narrow range through regulation of Ca metabolism in the intestine, kidney, and bone. Calcium is incorporated and resorbed from bone during bone remodeling via cellular processes as well as by exchange. Both routes contribute to calcium homeostasis. To assess the magnitude of bone turnover contribution to calcium homeostasis we labeled bone with a Ca tracer and measured Ca release following stimulation or suppression of bone resorption. Young growing male rats (n = 162) were dosed with 45Ca to label skeletal Ca. After a one-month period to allow the label to incorporate into the skeleton, rats were treated with a bone resorption antagonist (OPG), a bone resorption agonist (RANKL), or vehicle control (PBS). Serum and urine 45Ca and total Ca, and serum TRACP5b (a bone resorption biomarker), were monitored for 45 days following treatment. Tracer data were analyzed by a compartmental model using WinSAAM to quantify dynamic changes in Ca metabolism and identify sites of change following treatment. In RANKL treated rats, both serum 45Ca and serum TRACP5b were increased by >70% due to a 25-fold increase in bone resorption. In OPG treated rats, both serum 45Ca and serum TRACP5b were suppressed by >70% due to a 75% decrease in bone resorption, a 3-fold increase in bone formation, and a 50% increase in absorption. Because TRACP5b and 45Ca responded similarly, we conclude that Ca release from bone into serum occurs mostly via osteoclast-mediated bone resorption. However, because serum Ca concentration did not change with altered resorption in response to either RANKL or OPG treatment, we also conclude that serum Ca concentration under normal dietary conditions in young growing male rats is maintained by processes in addition to cellular bone resorption.


Subject(s)
Bone Resorption/blood , Calcium/blood , Growth and Development , Osteoprotegerin/metabolism , Animals , Body Weight/drug effects , Bone Resorption/urine , Calcium/urine , Male , Models, Biological , Osteoprotegerin/administration & dosage , Osteoprotegerin/pharmacology , RANK Ligand/administration & dosage , RANK Ligand/pharmacology , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/metabolism
7.
Calcif Tissue Int ; 101(6): 641-653, 2017 12.
Article in English | MEDLINE | ID: mdl-29038882

ABSTRACT

Etelcalcetide, a novel peptide agonist of the calcium-sensing receptor, prevents vascular calcification in a rat model of renal insufficiency with secondary hyperparathyroidism. Vascular calcification occurs frequently in patients with chronic kidney disease (CKD) and is a consequence of impaired mineral homeostasis and secondary hyperparathyroidism (SHPT). Etelcalcetide substantially lowers parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) levels in SHPT patients on hemodialysis. This study compared the effects of etelcalcetide and paricalcitol on vascular calcification in rats with adenine-induced CKD and SHPT. Uremia and SHPT were induced in male Wistar rats fed a diet supplemented with 0.75% adenine for 4 weeks. Rats were injected with vehicle, etelcalcetide, or paricalcitol for 4 weeks from the beginning of adenine diet. Rats fed an adenine-free diet were included as nonuremic controls. Similar reductions in plasma PTH and parathyroid chief cell proliferation were observed in both etelcalcetide- and paricalcitol-treated rats. Serum calcium and phosphorus were significantly lower in etelcalcetide-treated uremic rats and was unchanged in paricalcitol-treated rats. Both serum FGF23 and aortic calcium content were significantly lower in etelcalcetide-treated uremic rats compared with either vehicle- or paricalcitol-treated uremic rats. The degree of aortic calcium content for etelcalcetide-treated rats was similar to that in nonuremic controls and corroborated findings of lack of histologic aortic mineralization in those groups. In conclusion, etelcalcetide and paricalcitol similarly attenuated progression of SHPT in an adenine rat model of CKD. However, etelcalcetide differentially prevented vascular calcification, at least in part, due to reductions in serum FGF23, calcium, and phosphorus levels.


Subject(s)
Hyperparathyroidism, Secondary/complications , Peptides/pharmacology , Renal Insufficiency/complications , Vascular Calcification/etiology , Animals , Disease Models, Animal , Ergocalciferols/pharmacology , Male , Rats , Rats, Wistar
8.
Bone ; 105: 163-172, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28867373

ABSTRACT

Sustained elevation of parathyroid hormone (PTH) is catabolic to cortical bone, as evidenced by deterioration in bone structure (cortical porosity), and is a major factor for increased fracture risk in chronic kidney disease (CKD). Etelcalcetide (AMG 416), a novel peptide agonist of the calcium-sensing receptor, reduces PTH levels in subtotal nephrectomized (Nx) rats and in hemodialysis patients with secondary hyperparathyroidism (SHPT) in clinical studies; however, effects of etelcalcetide on bone have not been determined. In a rat model of established SHPT with renal osteodystrophy, etelcalcetide or vehicle was administered by subcutaneous (s.c.) injection to subtotal Nx rats with elevated PTH (>750pg/mL) once per day for 6weeks. Sham-operated rats receiving vehicle (s.c.) served as non-SHPT controls. Prior to treatment, significant increases in serum creatinine (2-fold), blood urea nitrogen (BUN, 3-fold), PTH (5-fold), fibroblast growth factor-23 (FGF23; 13-fold) and osteocalcin (12-fold) were observed in SHPT rats compared to non-SHPT controls. Elevations in serum creatinine and BUN were unaffected by treatment with vehicle or etelcalcetide. In contrast, etelcalcetide significantly decreased PTH, FGF23 and osteocalcin, whereas vehicle treatment did not. Cortical bone porosity increased and bone strength decreased in vehicle-treated SHPT rats compared to non-SHPT controls. Cortical bone structure improved and energy to failure was significantly greater in SHPT rats treated with etelcalcetide compared to vehicle. Mineralization lag time and marrow fibrosis were significantly reduced by etelcalcetide. In conclusion, etelcalcetide reduced bone turnover, attenuated mineralization defect and marrow fibrosis, and preserved cortical bone structure and bone strength by lowering PTH in subtotal Nx rats with established SHPT.


Subject(s)
Cortical Bone/physiopathology , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/physiopathology , Nephrectomy , Peptides/therapeutic use , Receptors, Calcium-Sensing/agonists , Animals , Biomechanical Phenomena/drug effects , Blood Urea Nitrogen , Calcium/blood , Cortical Bone/drug effects , Creatinine/blood , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Hyperparathyroidism, Secondary/blood , Hyperplasia , Kidney Function Tests , Male , Osteocalcin/blood , Parathyroid Glands/pathology , Parathyroid Hormone/blood , Peptides/pharmacology , Phosphorus/blood , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/blood
9.
J Bone Miner Res ; 32(4): 834-845, 2017 04.
Article in English | MEDLINE | ID: mdl-27505721

ABSTRACT

Results of prior studies suggest that fibroblast growth factor 21 (FGF21) may be involved in bone turnover and in the actions of peroxisome proliferator-activated receptor (PPAR) α and γ in mice. We have conducted independent studies to examine the effects of FGF21 on bone homeostasis and the role of FGF21 in PPARα and γ actions. High-fat-diet-induced obesity (DIO) mice were administered vehicle or recombinant human FGF21 (rhFGF21) intraperitoneally at 0 (vehicle), 0.1, 1, and 3 mg/kg daily for 2 weeks. Additional groups of DIO mice received water or 10 mg/kg rosiglitazone daily. Mice treated with rhFGF21 or rosiglitazone showed expected metabolic improvements in glucose, insulin, and lipid levels. However, bone loss was not detected in rhFGF21-treated mice by dual-energy X-ray absorptiometry (DXA), micro-CT, and histomorphometric analyses. Mineral apposition rate, a key bone formation parameter, was unchanged by rhFGF21, while significantly decreased by rosiglitazone in DIO mice. Bone resorption markers, OPG/RANKL mRNA expression, and histological bone resorption indices were unchanged by rhFGF21 or rosiglitazone. Bone marrow fat was unchanged by rhFGF21, while increased by rosiglitazone. Furthermore, FGF21 knockout mice did not show high bone mass phenotype. Treatment with PPARα or PPARγ agonists caused similar metabolic effects in FGF21 knockout and wild-type mice. These results contrast with previous findings and suggest that FGF21 is not critical for bone homeostasis or actions of PPARα and PPARγ. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Density , Fibroblast Growth Factors , Gene Expression Regulation/drug effects , Homeostasis , PPAR alpha , PPAR gamma , Animals , Bone Density/drug effects , Bone Density/genetics , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Humans , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/metabolism , Osteoprotegerin/biosynthesis , Osteoprotegerin/genetics , PPAR alpha/agonists , PPAR alpha/biosynthesis , PPAR alpha/genetics , PPAR gamma/agonists , PPAR gamma/biosynthesis , PPAR gamma/genetics , RANK Ligand/biosynthesis , RANK Ligand/genetics , Rosiglitazone , Thiazolidinediones/pharmacology
10.
J Bone Miner Res ; 30(9): 1627-40, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25727550

ABSTRACT

Osteonecrosis of the jaws (ONJ) is a significant complication of antiresorptive medications, such as bisphosphonates and denosumab. Antiresorptive discontinuation to promote healing of ONJ lesions remains highly controversial and understudied. Here, we investigated whether antiresorptive discontinuation alters ONJ features in mice, employing the potent bisphosphonate zoledronic acid (ZA) or the receptor activator of NF-κB ligand (RANKL) inhibitor OPG-Fc, utilizing previously published ONJ animal models. Mice were treated with vehicle (veh), ZA, or OPG-Fc for 11 weeks to induce ONJ, and antiresorptives were discontinued for 6 or 10 weeks. Maxillae and mandibles were examined by µCT imaging and histologically. ONJ features in ZA and OPG-Fc groups included periosteal bone deposition, empty osteocyte lacunae, osteonecrotic areas, and bone exposure, each of which substantially resolved 10 weeks after discontinuing OPG-Fc but not ZA. Full recovery of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclast numbers occurred after discontinuing OPG-Fc but not ZA. Our data provide the first experimental evidence demonstrating that discontinuation of a RANKL inhibitor, but not a bisphosphonate, reverses features of osteonecrosis in mice. It remains unclear whether antiresorptive discontinuation increases the risk of skeletal-related events in patients with bone metastases or fracture risk in osteoporosis patients, but these preclinical data may nonetheless help to inform discussions on the rationale for a "drug holiday" in managing the ONJ patient.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw/drug therapy , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Denosumab/pharmacology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Osteoclasts/metabolism , RANK Ligand/metabolism , Abscess , Acid Phosphatase/metabolism , Animals , Bone Resorption , Diphosphonates/therapeutic use , Disease Models, Animal , Immunoglobulin Fc Fragments/pharmacology , Isoenzymes/metabolism , Male , Mandible/diagnostic imaging , Maxilla/diagnostic imaging , Mice , Mice, Inbred C57BL , Osteoprotegerin/pharmacology , RANK Ligand/antagonists & inhibitors , Rats , Recombinant Fusion Proteins/pharmacology , Tartrate-Resistant Acid Phosphatase , X-Ray Microtomography , Zoledronic Acid
11.
Endocrinology ; 155(12): 4785-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25259718

ABSTRACT

The effects of up to 26 weeks of sclerostin antibody (Scl-Ab) treatment were investigated in ovariectomized (OVX) rats. Two months after surgery, 6-month-old osteopenic OVX rats were treated with vehicle or Scl-Ab (25 mg/kg, sc, one time per week) for 6, 12, or 26 weeks. In vivo dual-energy x-ray absorptiometry analysis demonstrated that the bone mineral density of lumbar vertebrae and femur-tibia increased progressively through 26 weeks of Scl-Ab treatment along with progressive increases in trabecular and cortical bone mass and bone strength at multiple sites. There was a strong correlation between bone mass and maximum load at lumbar vertebra, femoral neck, and diaphysis at weeks 6 and 26. Dynamic histomorphometric analysis showed that lumbar trabecular and tibial shaft endocortical and periosteal bone formation rates (BFR/BS) increased and peaked at week 6 with Scl-Ab-treatment; thereafter trabecular and endocortical BFR/BS gradually declined but remained significantly greater than OVX controls at week 26, whereas periosteal BFR/BS returned to OVX control levels at week 26. In the tibia metaphysis, trabecular BFR/BS in the Scl-Ab treated group remained elevated from week 6 to week 26. The osteoclast surface and eroded surface were significantly lower in Scl-Ab-treated rats than in OVX controls at all times. In summary, bone mass and strength increased progressively over 26 weeks of Scl-Ab treatment in adult OVX rats. The early gains were accompanied by increased cortical and trabecular bone formation and reduced osteoclast activity, whereas later gains were attributed to residual endocortical and trabecular osteoblast stimulation and persistently low osteoclast activity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone and Bones/drug effects , Osteoporosis/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Bone Density/drug effects , Bone Remodeling/drug effects , Drug Evaluation, Preclinical , Female , Genetic Markers , Ovariectomy , Random Allocation , Rats, Sprague-Dawley , X-Ray Microtomography
12.
Bone ; 67: 305-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25093263

ABSTRACT

Sclerostin (Scl) is an osteocyte protein that decreases bone formation, and its inhibition by neutralizing antibodies (Scl-Ab) increases bone formation, mass and strength. We investigated the effects of Scl-Ab in mature ovariectomized (OVX) rats with a mechanistic focus on longer-term responses of osteoclasts, osteoblasts and osteocytes. Four-month-old Sprague-Dawley rats had OVX or sham surgery. Two months later, sham controls received sc vehicle while OVX rats received vehicle (OVX-Veh) or Scl-Ab (25mg/kg) once weekly for 6 or 26weeks followed by necropsy (n=12/group). Terminal blood was collected for biochemistry, non-adherent marrow cells were harvested from femurs for ex vivo osteoclast formation assays, and vertebrae and tibiae were collected for dynamic histomorphometry and mRNA analyses. Scl-Ab treatment led to progressively thicker but fewer trabeculae in the vertebra, leading to increased trabecular bone volume and reduced trabecular surfaces. Scl-Ab also increased cortical bone volume in the tibia, via early periosteal expansion and progressive endocortical contraction. Scl-Ab significantly reduced parameters of bone resorption at week 6 relative to OVX-Veh controls, including reduced serum TRACP-5b, reduced capacity of marrow cells to form osteoclasts ex vivo, and >80% reductions in vertebral trabecular and tibial endocortical eroded surfaces. At week 26, serum TRACP-5b and ex vivo osteoclast formation were no longer reduced in the Scl-Ab group, but eroded surfaces remained >80% lower than in OVX-Veh controls without evidence for altered skeletal mRNA expression of opg or rankl. Scl-Ab significantly increased parameters of bone formation at week 6 relative to OVX-Veh controls, including increases in serum P1NP and osteocalcin, and increased trabecular, endocortical and periosteal bone formation rates (BFRs). At week 26, surface-referent trabecular BFR remained significantly increased in the Scl-Ab group versus OVX-Veh controls, but after adjusting for a reduced extent of trabecular surfaces, overall (referent-independent) trabecular BFR was no longer significantly elevated. Similarly, serum P1NP and osteocalcin were no longer significantly increased in the Scl-Ab group at week 26. Tibial endocortical and periosteal BFR were increased at week 6 in the Scl-Ab group versus OVX-Veh controls, while at week 26 only endocortical BFR remained increased. The Scl-Ab group exhibited significant increments in skeletal mRNA expression of several osteocyte genes, with sost showing the greatest induction in both the tibia and vertebra. We propose that Scl-Ab administration, and/or the gains in bone volume that result, may have increased osteocytic expression of Scl as a possible means of regulating gains in bone mass.


Subject(s)
Antibodies/pharmacology , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Animals , Antibodies/administration & dosage , Bone Density/drug effects , Female , Osteoclasts/drug effects , Osteogenesis/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley
13.
J Orthop Res ; 32(2): 197-203, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24600701

ABSTRACT

Systemic administration of a sclerostin neutralizing antibody (Scl-Ab) has been shown to enhance fracture callus density and strength in several animal models. In order to further evaluate the potential of Scl-Ab to improve healing in a bone defect model,we evaluated Scl-Ab in a 3mm femoral defect in young male outbred rats. Scl-Ab was given either continuously for 6 or 12 weeks after surgery or with 2 weeks of delay for 10 weeks. Bone formation was assessed by radiographs, µ-CT, and histology. Complete bony union was achieved in only a few defects after 12 weeks of healing (Scl-Ab treated 5/30, vehicle treated 1/15). µ-CT evaluation demonstrated a significant increase in the BV/TV in the defect in the delayed treatment group (65%, p<0.05), but a non-significant increase in the continuous group (35%, p = 0.11) compared to control. However, both regimens induced an anabolic response in the bone proximal and distal to the defect and in the un-operated femurs. We demonstrate that treatment with Scl-Ab can enhance bone repair in a bone defect and in the surrounding host bone, but lacks the osteoinductive activity to heal it. This agent seems to be most effective in bone repair scenarios where there is cortical integrity.


Subject(s)
Bone Morphogenetic Proteins/immunology , Fracture Healing/drug effects , Genetic Markers/immunology , Osteogenesis/drug effects , Animals , Antibodies, Neutralizing , Femoral Fractures , Femur/drug effects , Male , Rats , Rats, Sprague-Dawley , X-Ray Microtomography
14.
Ann Rheum Dis ; 72(10): 1732-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23666928

ABSTRACT

OBJECTIVE: To test whether inhibition of sclerostin by a targeted monoclonal antibody (Scl-Ab) protects from bone and cartilage damage in inflammatory arthritis. Sclerostin is a potent inhibitor of bone formation and may be responsible for the low level of bone repair in patients with rheumatoid arthritis. METHODS: Human tumour necrosis factor transgenic mice (hTNFtg mice) developing inflammatory arthritis and local and bone loss were administered either vehicle, anti-TNF antibody, Scl-Ab, or a combination of both agents. Inflammation, systemic and periarticular bone loss, bone erosion and cartilage damage were evaluated at baseline (week 8) and after 3 weeks of treatment by clinical assessment, micro-CT and histology. RESULTS: Scl-Ab did not affect joint swelling or synovitis. Systemic bone loss in the spine and periarticular bone loss in the proximal tibia were completely blocked and partially reversed by inhibition of sclerostin but not by inhibition of TNF. Moreover, Scl-Ab completely arrested the progression of bone erosion in hTNFtg mice and in combination with TNF inhibition even led to significant regression of cortical bone erosions. Protective effects of Scl-Ab were also observed for the articular cartilage. CONCLUSIONS: These data suggest that sclerostin inhibition is a powerful tool to enhance bone repair in inflammatory arthritis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/complications , Bone Diseases, Metabolic/drug therapy , Glycoproteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/pathology , Bone Regeneration/drug effects , Cartilage Diseases/pathology , Cartilage Diseases/prevention & control , Cartilage, Articular/pathology , Disease Progression , Drug Evaluation, Preclinical/methods , Female , Intercellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Tumor Necrosis Factor-alpha/antagonists & inhibitors
15.
J Bone Miner Res ; 28(4): 865-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23109229

ABSTRACT

Sclerostin, a product of the SOST gene produced mainly by osteocytes, is a potent negative regulator of bone formation that appears to be responsive to mechanical loading, with SOST expression increasing following mechanical unloading. We tested the ability of a murine sclerostin antibody (SclAbII) to prevent bone loss in adult mice subjected to hindlimb unloading (HLU) via tail suspension for 21 days. Mice (n = 11-17/group) were assigned to control (CON, normal weight bearing) or HLU and injected with either SclAbII (subcutaneously, 25 mg/kg) or vehicle (VEH) twice weekly. SclAbII completely inhibited the bone deterioration due to disuse, and induced bone formation such that bone properties in HLU-SclAbII were at or above values of CON-VEH mice. For example, hindlimb bone mineral density (BMD) decreased -9.2% ± 1.0% in HLU-VEH, whereas it increased 4.2% ± 0.7%, 13.1% ± 1.0%, and 30.6% ± 3.0% in CON-VEH, HLU-SclAbII, and CON-SclAbII, respectively (p < 0.0001). Trabecular bone volume, assessed by micro-computed tomography (µCT) imaging of the distal femur, was lower in HLU-VEH versus CON-VEH (p < 0.05), and was 2- to 3-fold higher in SclAbII groups versus VEH (p < 0.001). Midshaft femoral strength, assessed by three-point bending, and distal femoral strength, assessed by micro-finite element analysis (µFEA), were significantly higher in SclAbII versus VEH-groups in both loading conditions. Serum sclerostin was higher in HLU-VEH (134 ± 5 pg/mL) compared to CON-VEH (116 ± 6 pg/mL, p < 0.05). Serum osteocalcin was decreased by hindlimb suspension and increased by SclAbII treatment. Interestingly, the anabolic effects of sclerostin inhibition on some bone outcomes appeared to be enhanced by normal mechanical loading. Altogether, these results confirm the ability of SclAbII to abrogate disuse-induced bone loss and demonstrate that sclerostin antibody treatment increases bone mass by increasing bone formation in both normally loaded and underloaded environments.


Subject(s)
Antibodies/pharmacology , Bone and Bones/pathology , Glycoproteins/immunology , Adaptor Proteins, Signal Transducing , Animals , Biomarkers/metabolism , Biomechanical Phenomena/drug effects , Body Weight/drug effects , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/physiopathology , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Femur/physiopathology , Finite Element Analysis , Glycoproteins/blood , Hindlimb Suspension , Intercellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Muscles/drug effects , Muscles/pathology , Organ Size/drug effects , Weight-Bearing/physiology , X-Ray Microtomography
16.
Bone ; 50(1): 209-17, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22075208

ABSTRACT

Sclerostin, the Wnt signaling antagonist encoded by the Sost gene, is secreted by osteocytes and inhibits bone formation by osteoblasts. Mechanical stimulation reduces sclerostin expression, suggesting that osteocytes might coordinate the osteogenic response to mechanical force by locally unleashing Wnt signaling. To investigate whether sclerostin downregulation is a pre-requisite for load-induced bone formation, we conducted experiments in transgenic mice (TG) engineered to maintain high levels of SOST expression during mechanical loading. This was accomplished by introducing a human SOST transgene driven by the 8 kb fragment of the DMP1 promoter that also provided osteocyte specificity of the transgene. Right ulnae were subjected to in vivo cyclic axial loading at equivalent strains for 1 min/day at 2 Hz; left ulnae served as internal controls. Endogenous murine Sost mRNA expression measured 24 h after 1 loading bout was decreased by about 50% in TG and wild type (WT) littermates. In contrast, human SOST, only expressed in TG mice, remained high after loading. Mice were loaded on 3 consecutive days and bone formation was quantified 16 days after initiation of loading. Periosteal bone formation in control ulnae was similar in WT and TG mice. Loading induced the expected strain-dependent increase in bone formation in WT mice, resulting from increases in both mineralizing surface (MS/BS) and mineral apposition rate (MAR). In contrast, load-induced bone formation was reduced by 70-85% in TG mice, due to lower MS/BS and complete inhibition of MAR. Moreover, Wnt target gene expression induced by loading in WT mice was absent in TG mice. Thus, downregulation of Sost/sclerostin in osteocytes is an obligatory step in the mechanotransduction cascade that activates Wnt signaling and directs osteogenesis to where bone is structurally needed.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Glycoproteins/metabolism , Osteogenesis/physiology , Signal Transduction/physiology , Weight-Bearing/physiology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Down-Regulation , Female , Genetic Markers/genetics , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Male , Mechanotransduction, Cellular/physiology , Mice , Mice, Transgenic , Osteocytes/physiology , Stress, Mechanical , Transgenes
17.
Bone ; 50(3): 628-37, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154841

ABSTRACT

We examined age-related changes in biochemical markers and regulators of osteoblast and osteoclast activity in C57BL/6 mice to assess their utility in explaining age-related changes in bone. Several recently discovered regulators of osteoclasts and osteoblasts were also measured to assess concordance between their systemic levels versus their levels in marrow plasma, to which bone cells are directly exposed. MicroCT of 6-, 12-, and 24-month-old mice indicated an early age-related loss of trabecular bone volume and surface, followed by endocortical bone loss and periosteal expansion. Trabecular bone loss temporally correlated with reductions in biomarkers of bone formation and resorption in both peripheral blood and bone marrow. Endocortical bone loss and periosteal bone gain were not reflected in these protein biomarkers, but were well correlated with increased expression of osteocalcin, rank, tracp5b, and cathepsinK in RNA extracted from cortical bone. While age-related changes in bone turnover markers remained concordant in blood versus marrow, aging led to divergent changes in blood versus marrow for the bone cell regulators RANKL, OPG, sclerostin, DKK1, and serotonin. Bone expression of runx2 and osterix increased progressively with aging and was associated with an increase in the number of osteoprogenitors and osteoclast precursors. In summary, levels of biochemical markers of bone turnover in blood and bone marrow plasma were predictive of an age-related loss of trabecular surfaces in adult C57BL/6 mice, but did not predict gains in cortical surfaces resulting from cortical expansion. Unlike these turnover markers, a panel of bone cell regulatory proteins exhibited divergent age-related changes in marrow versus peripheral blood, suggesting that their circulating levels may not reflect local levels to which osteoclasts and osteoblasts are directly exposed.


Subject(s)
Aging/metabolism , Bone Remodeling/physiology , Bone and Bones/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Animals , Biomarkers/blood , Bone Marrow/metabolism , Cells, Cultured , Male , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism
18.
Endocrinology ; 152(9): 3312-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21733832

ABSTRACT

Clinical studies have revealed a blunting of the bone anabolic effects of parathyroid hormone treatment in osteoporotic patients in the setting of pre- or cotreatment with the antiresorptive agent alendronate (ALN). Sclerostin monoclonal antibody (Scl-Ab) is currently under clinical investigation as a new potential anabolic therapy for postmenopausal osteoporosis. The purpose of these experiments was to examine the influence of pretreatment or cotreatment with ALN on the bone anabolic actions of Scl-Ab in ovariectomized (OVX) rats. Ten-month-old osteopenic OVX rats were treated with ALN or vehicle for 6 wk, before the start of Scl-Ab treatment. ALN-pretreated OVX rats were switched to Scl-Ab alone or to a combination of ALN and Scl-Ab for another 6 wk. Vehicle-pretreated OVX rats were switched to Scl-Ab or continued on vehicle to serve as controls. Scl-Ab treatment increased areal bone mineral density, volumetric bone mineral density, trabecular and cortical bone mass, and bone strength similarly in OVX rats pretreated with ALN or vehicle. Serum osteocalcin and bone formation rate on trabecular, endocortical, and periosteal surfaces responded similarly to Scl-Ab in ALN or vehicle-pretreated OVX rats. Furthermore, cotreatment with ALN did not have significant effects on the increased bone formation, bone mass, and bone strength induced by Scl-Ab in the OVX rats that were pretreated with ALN. These results indicate that the increases in bone formation, bone mass, and bone strength with Scl-Ab treatment were not affected by pre- or cotreatment with ALN in OVX rats with established osteopenia.


Subject(s)
Alendronate/pharmacology , Antibodies, Monoclonal/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Osteogenesis/drug effects , Acid Phosphatase/blood , Alendronate/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Bone Density/immunology , Bone Density Conservation Agents/therapeutic use , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/immunology , Bone and Bones/drug effects , Bone and Bones/immunology , Disease Models, Animal , Female , Isoenzymes/blood , Osteocalcin/blood , Osteogenesis/immunology , Ovariectomy , Rats , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase
19.
J Bone Miner Res ; 26(11): 2610-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21773994

ABSTRACT

The physiological role of Dickkopf-1 (Dkk1) during postnatal bone growth in rodents and in adult rodents was examined utilizing an antibody to Dkk1 (Dkk1-Ab) that blocked Dkk1 binding to both low density lipoprotein receptor-related protein 6 (LRP6) and Kremen2, thereby preventing the Wnt inhibitory activity of Dkk1. Treatment of growing mice and rats with Dkk1-Ab resulted in a significant increase in bone mineral density because of increased bone formation. In contrast, treatment of adult ovariectomized rats did not appreciably impact bone, an effect that was associated with decreased Dkk1 expression in the serum and bone of older rats. Finally, we showed that Dkk1 plays a prominent role in adult bone by mediating fracture healing in adult rodents. These data suggest that, whereas Dkk1 significantly regulates bone formation in younger animals, its role in older animals is limited to pathologies that lead to the induction of Dkk1 expression in bone and/or serum, such as traumatic injury.


Subject(s)
Aging/metabolism , Bone and Bones/injuries , Bone and Bones/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteogenesis/physiology , Aging/drug effects , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/pharmacology , Bone Density/drug effects , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cell Line , Estrogens/deficiency , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Fracture Healing/drug effects , Humans , Intercellular Signaling Peptides and Proteins/blood , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Male , Mice , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects , X-Ray Microtomography
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...