Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Physiol Rep ; 9(21): e15081, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34755490

RESUMEN

Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Orthomyxoviridae/patogenicidad , Neumonía Viral/tratamiento farmacológico , Angiopoyetina 2/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Células Cultivadas , Citocinas/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Neumonía Viral/metabolismo , Neumonía Viral/virología , Receptor TIE-2/metabolismo , Carga Viral
2.
Sci Rep ; 11(1): 11839, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088950

RESUMEN

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.


Asunto(s)
Perfilación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Femenino , Fibroblastos/metabolismo , Redes Reguladoras de Genes , Atrios Cardíacos/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/fisiopatología , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Necrosis , Estrés Oxidativo , Pronóstico , Transducción de Señal , Volumen Sistólico/fisiología , Transcripción Genética , Transcriptoma , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda/fisiología
3.
PLoS One ; 15(8): e0231234, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32804947

RESUMEN

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.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Síndrome Metabólico/metabolismo , Animales , Biomarcadores/metabolismo , Corazón/fisiología , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/fisiopatología , Riñón/metabolismo , Masculino , Síndrome Metabólico/complicaciones , Miocardio/metabolismo , Obesidad/complicaciones , Ratas , Ratas Endogámicas , Ratas Zucker , Volumen Sistólico/fisiología , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/fisiología
4.
Bone ; 116: 162-170, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30077758

RESUMEN

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.


Asunto(s)
Resorción Ósea/sangre , Calcio/sangre , Crecimiento y Desarrollo , Osteoprotegerina/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Resorción Ósea/orina , Calcio/orina , Masculino , Modelos Biológicos , Osteoprotegerina/administración & dosificación , Osteoprotegerina/farmacología , Ligando RANK/administración & dosificación , Ligando RANK/farmacología , Ratas Sprague-Dawley , Fosfatasa Ácida Tartratorresistente/metabolismo
5.
Calcif Tissue Int ; 101(6): 641-653, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29038882

RESUMEN

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.


Asunto(s)
Hiperparatiroidismo Secundario/complicaciones , Péptidos/farmacología , Insuficiencia Renal/complicaciones , Calcificación Vascular/etiología , Animales , Modelos Animales de Enfermedad , Ergocalciferoles/farmacología , Masculino , Ratas , Ratas Wistar
6.
Bone ; 105: 163-172, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28867373

RESUMEN

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.


Asunto(s)
Hueso Cortical/fisiopatología , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hiperparatiroidismo Secundario/fisiopatología , Nefrectomía , Péptidos/uso terapéutico , Receptores Sensibles al Calcio/agonistas , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Calcio/sangre , Hueso Cortical/efectos de los fármacos , Creatinina/sangre , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Hiperparatiroidismo Secundario/sangre , Hiperplasia , Pruebas de Función Renal , Masculino , Osteocalcina/sangre , Glándulas Paratiroides/patología , Hormona Paratiroidea/sangre , Péptidos/farmacología , Fósforo/sangre , Ratas Sprague-Dawley , Fosfatasa Ácida Tartratorresistente/sangre
7.
J Bone Miner Res ; 32(4): 834-845, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27505721

RESUMEN

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.


Asunto(s)
Densidad Ósea , Factores de Crecimiento de Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , PPAR alfa , PPAR gamma , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Insulina/genética , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/metabolismo , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , PPAR alfa/agonistas , PPAR alfa/biosíntesis , PPAR alfa/genética , PPAR gamma/agonistas , PPAR gamma/biosíntesis , PPAR gamma/genética , Ligando RANK/biosíntesis , Ligando RANK/genética , Rosiglitazona , Tiazolidinedionas/farmacología
8.
Nat Commun ; 7: 11505, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27230681

RESUMEN

Inhibition of the Wnt antagonist sclerostin increases bone mass in patients with osteoporosis and in preclinical animal models. Here we show increased levels of the Wnt antagonist Dickkopf-1 (DKK-1) in animals treated with sclerostin antibody, suggesting a negative feedback mechanism that limits Wnt-driven bone formation. To test our hypothesis that co-inhibition of both factors further increases bone mass, we engineer a first-in-class bispecific antibody with single residue pair mutations in the Fab region to promote efficient and stable cognate light-heavy chain pairing. We demonstrate that dual inhibition of sclerostin and DKK-1 leads to synergistic bone formation in rodents and non-human primates. Furthermore, by targeting distinct facets of fracture healing, the bispecific antibody shows superior bone repair activity compared with monotherapies. This work supports the potential of this agent both for treatment and prevention of fractures and offers a promising therapeutic approach to reduce the burden of low bone mass disorders.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Fracturas Óseas/tratamiento farmacológico , Fracturas Óseas/fisiopatología , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Densidad Ósea , Modelos Animales de Enfermedad , Femenino , Fracturas Óseas/genética , Fracturas Óseas/metabolismo , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Macaca fascicularis , Masculino , Ratones , Ratones Noqueados , Osteogénesis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Vía de Señalización Wnt/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
9.
Bone ; 75: 96-104, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25701138

RESUMEN

In addition to its thoroughly investigated role in bone formation, the osteoblast master transcription factor RUNX2 also promotes osteoclastogenesis and bone resorption. Here we demonstrate that 17ß-estradiol (E2), strongly inhibits RUNX2-mediated osteoblast-driven osteoclastogenesis in co-cultures. Towards deciphering the underlying mechanism, we induced premature expression of RUNX2 in primary murine pre-osteoblasts, which resulted in robust differentiation of co-cultured splenocytes into mature osteoclasts. This was attributable to RUNX2-mediated increase in RANKL secretion, determined by ELISA, as well as to RUNX2-mediated increase in RANKL association with the osteoblast membrane, demonstrated using confocal fluorescence microscopy. The increased association with the osteoblast membrane was recapitulated by transiently expressed GFP-RANKL. E2 abolished the RUNX2-mediated increase in membrane-associated RANKL and GFP-RANKL, as well as the concomitant osteoclastogenesis. RUNX2-mediated RANKL cellular redistribution was attributable in part to a decrease in Opg expression, but E2 did not influence Opg expression either in the presence or absence of RUNX2. Diminution of RUNX2-mediated osteoclastogenesis by E2 occurred regardless of whether the pre-osteoclasts were derived from wild type or estrogen receptor alpha (ERα)-knockout mice, suggesting that activated ERα inhibited osteoblast-driven osteoclastogenesis by acting in osteoblasts, possibly targeting RUNX2. Indeed, microarray analysis demonstrated global attenuation of the RUNX2 response by E2, including abrogation of Pstpip2 expression, which likely plays a critical role in membrane trafficking. Finally, the selective ER modulators (SERMs) tamoxifen and raloxifene mimicked E2 in abrogating the stimulatory effect of osteoblastic RUNX2 on osteoclast differentiation in the co-culture assay. Thus, E2 antagonizes RUNX2-mediated RANKL trafficking and subsequent osteoclastogenesis. Targeting RUNX2 and/or downstream mechanisms that regulate RANKL trafficking may lead to the development of improved SERMs and possibly non-hormonal therapeutic approaches to high turnover bone disease.


Asunto(s)
Resorción Ósea/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Estrógenos/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Animales , Western Blotting , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/citología , Osteoclastos/citología , Reacción en Cadena de la Polimerasa
10.
Endocrinology ; 155(12): 4785-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25259718

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Huesos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Densidad Ósea/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Marcadores Genéticos , Ovariectomía , Distribución Aleatoria , Ratas Sprague-Dawley , Microtomografía por Rayos X
11.
Bone ; 67: 305-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25093263

RESUMEN

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.


Asunto(s)
Anticuerpos/farmacología , Proteínas Morfogenéticas Óseas/inmunología , Marcadores Genéticos/inmunología , Animales , Anticuerpos/administración & dosificación , Densidad Ósea/efectos de los fármacos , Femenino , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ovariectomía , Ratas , Ratas Sprague-Dawley
12.
J Bone Miner Res ; 29(5): 1158-69, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24127173

RESUMEN

Receptor activator of NF-κB ligand (RANKL) plays a key role in osteoclast-induced bone resorption across a range of degenerative bone diseases, and its specific inhibition has been recently approved as a treatment for women with postmenopausal osteoporosis at high or increased risk of fracture in the United States and globally. In the present study, we generated transgenic mice (TghuRANKL) carrying the human RANKL (huRANKL) genomic region and achieved a physiologically relevant pattern of RANKL overexpression in order to establish novel genetic models for assessing skeletal and extraskeletal pathologies associated with excessive RANKL and for testing clinical therapeutic candidates that inhibit human RANKL. TghuRANKL mice of both sexes developed early-onset bone loss, and the levels of huRANKL expression were correlated with bone resorption and disease severity. Low copy Tg5516 mice expressing huRANKL at low levels displayed a mild osteoporotic phenotype as shown by trabecular bone loss and reduced biomechanical properties. Notably, overexpression of huRANKL, in the medium copy Tg5519 line, resulted in severe early-onset osteoporosis characterized by lack of trabecular bone, destruction of the growth plate, increased osteoclastogenesis, bone marrow adiposity, increased bone remodeling, and severe cortical bone porosity accompanied by decreased bone strength. An even more severe skeletal phenotype developed in the high copy Tg5520 founder with extensive soft tissue calcification. Model validation was further established by evidence that denosumab, an antibody that inhibits human but not murine RANKL, fully corrected the hyper-resorptive and osteoporotic phenotypes of Tg5519 mice. Furthermore, overexpression of huRANKL rescued osteopetrotic phenotypes of RANKL-defective mice. These novel huRANKL transgenic models of osteoporosis represent an important advance for understanding the pathogenesis and treatment of high-turnover bone diseases and other disease states caused by excessive RANKL.


Asunto(s)
Regulación de la Expresión Génica , Osteoporosis/genética , Osteoporosis/metabolismo , Ligando RANK/biosíntesis , Animales , Calcinosis/genética , Calcinosis/metabolismo , Calcinosis/patología , Modelos Animales de Enfermedad , Femenino , Placa de Crecimiento/metabolismo , Placa de Crecimiento/patología , Humanos , Ratones , Ratones Transgénicos , Modelos Genéticos , Osteoclastos/patología , Osteopetrosis/genética , Osteopetrosis/metabolismo , Osteopetrosis/patología , Osteoporosis/patología , Ligando RANK/genética
13.
J Bone Miner Res ; 28(3): 627-38, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23109114

RESUMEN

Type 2 diabetes mellitus results in increased risk of fracture and delayed fracture healing. ZDF fa/fa rats are an established model of type 2 diabetes mellitus with low bone mass and delayed bone healing. We tested whether a sclerostin-neutralizing antibody (Scl-AbVI) would reverse the skeletal deficits of diabetic ZDF rats. Femoral defects of 3 mm were created in 11-week-old diabetic ZDF fa/fa and nondiabetic ZDF +/+ rats and stabilized by an internal plate. Saline or 25 mg/kg Scl-AbVI was administered subcutaneously (s.c.) twice weekly for 12 weeks (n = 9-10/group). Bone mass and strength were assessed using pQCT, micro-computed tomography (µCT), and biomechanical testing. Bone histomorphometry was used to assess bone formation, and the filling of the bone defect was analyzed by µCT. Diabetic rats displayed lower spinal and femoral bone mass compared to nondiabetic rats, and Scl-AbVI treatment significantly enhanced bone mass of the femur and the spine of diabetic rats (p < 0.0001). Scl-AbVI also reversed the deficit in bone strength in the diabetic rats, with 65% and 89% increases in maximum load at the femoral shaft and neck, respectively (p < 0.0001). The lower bone mass in diabetic rats was associated with a 65% decrease in vertebral bone formation rate, which Scl-AbVI increased by sixfold, consistent with a pronounced anabolic effect. Nondiabetic rats filled 57% of the femoral defect, whereas diabetic rats filled only 21% (p < 0.05). Scl-AbVI treatment increased defect regeneration by 47% and 74%, respectively (p < 0.05). Sclerostin antibody treatment reverses the adverse effects of type 2 diabetes mellitus on bone mass and strength, and improves bone defect regeneration in rats.


Asunto(s)
Anticuerpos/farmacología , Proteínas Morfogenéticas Óseas/inmunología , Huesos/efectos de los fármacos , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Marcadores Genéticos/inmunología , Tamaño de los Órganos/efectos de los fármacos , Animales , Fenómenos Biomecánicos , Western Blotting , Densidad Ósea , Huesos/fisiopatología , Masculino , Ratas , Regeneración
14.
Bone ; 49(6): 1131-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21890009

RESUMEN

Sclerostin is a secreted inhibitor of Wnt signaling and plays an essential role in the regulation of bone mass. The expression of sclerostin is largely restricted to osteocytes although its mode of transcriptional regulation is not well understood. We observed regulated expression of sclerostin mRNA and protein that was directly correlated with the mineralization response in cultured human Saos-2 osteosarcoma cells and rat primary calvarial cells. Sclerostin mRNA and protein levels were increased following treatment of cells with BMP2, BMP4 and BMP7. Analysis of deletion mutants from the -7.4 kb upstream region of the human sclerostin promoter did not reveal any specific regions that were responsive to BMPs, Wnt3a, PTH, TGFß1 or Activin A in Saos-2 cells. The downstream ECR5 element did not show enhancer activity in Saos-2 cells and also was not affected when Saos-2 cells were treated with BMPs or PTH. Genome-wide microarray analysis of Saos-2 cells treated with BMP2 showed significant changes in expression of several transcription factors with putative consensus DNA binding sites in the region of the sclerostin promoter. However, whereas most factors tested showed either a range of inhibitory activity (DLX family, MSX2, HEY1, SMAD6/7) or lack of activity on the sclerostin promoter including SMAD9, only MEF2B showed a positive effect on both the promoter and ECR5 element. These results suggest that the dramatic induction of sclerostin gene expression by BMPs in Saos-2 cells occurs indirectly and is associated with late stage differentiation of osteoblasts and the mineralization process.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Elementos de Facilitación Genéticos/genética , Marcadores Genéticos/genética , Osteosarcoma/genética , Regiones Promotoras Genéticas/genética , Activinas/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteosarcoma/patología , Hormona Paratiroidea/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Proteína Wnt3A/farmacología
15.
J Bone Miner Res ; 26(11): 2610-21, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21773994

RESUMEN

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.


Asunto(s)
Envejecimiento/metabolismo , Huesos/lesiones , Huesos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteogénesis/fisiología , Envejecimiento/efectos de los fármacos , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/farmacología , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/sangre , Enfermedades Óseas Metabólicas/fisiopatología , Huesos/diagnóstico por imagen , Huesos/patología , Línea Celular , Estrógenos/deficiencia , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Curación de Fractura/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Masculino , Ratones , Osteogénesis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Microtomografía por Rayos X
16.
J Bone Miner Res ; 26(5): 1012-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542004

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Densidad Ósea/efectos de los fármacos , Fracturas del Fémur/fisiopatología , Curación de Fractura/efectos de los fármacos , Glicoproteínas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Diáfisis/efectos de los fármacos , Diáfisis/patología , Diáfisis/fisiopatología , Modelos Animales de Enfermedad , Fémur/efectos de los fármacos , Fémur/patología , Fémur/fisiopatología , Peroné/efectos de los fármacos , Peroné/patología , Peroné/fisiopatología , Glicoproteínas/inmunología , Péptidos y Proteínas de Señalización Intercelular , Macaca fascicularis , Masculino , Tamaño de los Órganos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteotomía , Ratas , Ratas Sprague-Dawley
17.
Bioorg Med Chem Lett ; 19(17): 4924-8, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19648005

RESUMEN

A screening campaign of a diverse collection of approximately 250,000 small molecule compounds was performed to identify inhibitors of proline-rich tyrosine kinase 2 (Pyk2) with potential osteogenic activity in osteoblast cells. Compounds were prioritized based on selectivity following a counter-screen against focal adhesion kinase (FAK), a closely related kinase. 4-Amino and 5-aryl substituted pyridinone series were identified that showed strong biochemical potency against Pyk2 and up to 3700-fold selectivity over FAK. Modeling analysis suggested that structural differences in the substrate binding cleft could explain the high selectivity of these chemical series against FAK. Representative compounds from each series showed inhibition of Pyk2 autophosphorylation in 293T cells (IC(50) approximately 0.11 microM), complete inhibition of endogenous Pyk2 in A7r5 cells and increased levels of osteogenic markers in MC3T3 osteoblast cells (EC(50)'s approximately 0.01 microM). These results revealed a new class of compounds with osteogenic-inducing activity in osteoblast cells and a starting point for the development of more potent and selective Pyk2 inhibitors.


Asunto(s)
Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Osteoblastos/enzimología , Inhibidores de Proteínas Quinasas/química , Piridonas/química , Animales , Sitios de Unión , Línea Celular , Simulación por Computador , Quinasa 2 de Adhesión Focal/metabolismo , Humanos , Ratones , Osteoblastos/efectos de los fármacos , Fosforilación , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/síntesis química , Piridonas/farmacología , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
18.
J Bone Miner Res ; 24(9): 1552-64, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19419305

RESUMEN

A large genome-wide, recessive, N-ethyl-N-nitrosourea (ENU)-induced mutagenesis screen was performed on a mixed C57BL/6J and C3H.SW-H2/SnJ mouse background to identify genes regulating bone mass. Approximately 6500 male and female G(3) hybrid mice were phenotyped at 8 and 10 wk of age by DXA analysis for evidence of changes in unadjusted or body weight-adjusted BMD or BMC. Phenodeviant lines were identified based on statistical criteria that included a false discovery rate (FDR) <20% and Z-score >2.8. Genome-wide mapping scans were initiated on 22 lines, with evidence of high or low BMD or BMC that deviated by approximately -30% to +50% from the means. Several lines were discontinued as showing lack of heritability, but two heritable lines were identified with narrow chromosomal regions that allowed sequencing of potential mutant candidate genes. Novel mutations were identified in the Enpp1 (C397S) gene on chromosome 10 (line 4482) and the Ptpn6 (I482F) gene on chromosome 6 (line 4489) that were both associated with low bone mass. In addition, the phenotype of the Enpp1 mice showed a striking joint disease and calcification of blood vessels including the aorta, myocardium, and renal arteries and capillaries. These results support a role for the Enpp1 gene in the pathogenesis associated with mineralization of articular cartilage and vascular calcification. This work confirms the utility of the chemical mutagenesis approach for identification of potential disease genes and confirms the role of Enpp1 and Ptpn6 in regulating mineralization and skeletal bone mass.


Asunto(s)
Densidad Ósea/genética , Calcinosis/genética , Artropatías/genética , Hidrolasas Diéster Fosfóricas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Pirofosfatasas/genética , Enfermedades Vasculares/genética , Absorciometría de Fotón , Animales , Secuencia de Bases , Mapeo Cromosómico , Cartilla de ADN , Etilnitrosourea/toxicidad , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutagénesis , Mutágenos/toxicidad , Reacción en Cadena de la Polimerasa
19.
Bioorg Med Chem Lett ; 19(5): 1442-5, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19179072

RESUMEN

A chemical screen of 45,000 compounds from a diverse collection led to the identification of two series of small molecules with potent osteogenic activity in mouse MC3T3-E1 osteoblast cells. The first chemical group was characterized by an amino benzothiazole core (AMG0892 series) and the second group by a naphthyl amide core (AMG0309 series). Using alkaline phosphatase (ALP), osteocalcin (OCL) and calcium as markers of osteoblast differentiation and mineralization, both chemical series showed EC(50)s in the 0.01-0.2 microM range and were consistent for all three markers. Compounds inhibited cell proliferation, had no effect on apoptosis and showed evidence for CREB pathway activity. The present compounds represent some of the most potent osteogenic small molecules reported to date and provide new tools for elucidating signaling mechanisms in osteoblasts.


Asunto(s)
Benzotiazoles/química , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Células 3T3 , Anabolizantes/química , Anabolizantes/farmacología , Animales , Benzotiazoles/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos C3H , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Relación Estructura-Actividad
20.
Development ; 131(1): 229-40, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14660436

RESUMEN

We have identified a novel chordin-like protein, CHL2, which is structurally most homologous to CHL/neuralin/ventroptin. When injected into Xenopus embryos, CHL2 RNA induced a secondary axis. Recombinant CHL2 protein interacted directly with BMPs in a competitive manner to prevent binding to the type I BMP receptor ectodomain, and inhibited BMP-dependent induction of alkaline phosphatase in C2C12 cells. Thus, CHL2 behaves as a secreted BMP-binding inhibitor. In situ hybridization revealed that CHL2 expression is restricted to chondrocytes of various developing joint cartilage surfaces and connective tissues in reproductive organs. Adult mesenchymal progenitor cells expressed CHL2, and its levels decreased during chondrogenic differentiation. Addition of CHL2 protein to a chondrogenic culture system reduced cartilage matrix deposition. Consistently, CHL2 transcripts were weakly detected in normal adult joint cartilage. However, CHL2 expression was upregulated in middle zone chondrocytes in osteoarthritic joint cartilage (where hypertrophic markers are induced). CHL2 depressed chondrocyte mineralization when added during the hypertrophic differentiation of cultured hyaline cartilage particles. Thus, CHL2 may play negative roles in the (re)generation and maturation of articular chondrocytes in the hyaline cartilage of both developing and degenerated joints.


Asunto(s)
Proteínas Portadoras/genética , Cartílago/embriología , Condrocitos/fisiología , Osteoartritis/genética , Xenopus/embriología , Fosfatasa Alcalina/biosíntesis , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Proteínas Morfogenéticas Óseas/farmacología , Proteínas Portadoras/metabolismo , Secuencia Conservada , Cartilla de ADN , Embrión no Mamífero/fisiología , Proteínas de la Matriz Extracelular , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Placenta/fisiología , Embarazo , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA