Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 9: 515, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29881353

RESUMEN

Degenerative myopathies typically display a decline in satellite cells coupled with a replacement of muscle fibers by fat and fibrosis. During this pathological remodeling, satellite cells are present at lower numbers and do not display a proper regenerative function. Whether a decline in satellite cells directly contributes to disease progression or is a secondary result is unknown. In order to dissect these processes, we used a genetic model to reduce the satellite cell population by ~70-80% which leads to a nearly complete loss of regenerative potential. We observe that while no overt tissue damage is observed following satellite cell depletion, muscle fibers atrophy accompanied by changes in the stem cell niche cellular composition. Treatment of these mice with an Activin receptor type-2B (AcvR2B) pathway blocker reverses muscle fiber atrophy as expected, but also restores regenerative potential of the remaining satellite cells. These findings demonstrate that in addition to controlling fiber size, the AcvR2B pathway acts to regulate the muscle stem cell niche providing a more favorable environment for muscle regeneration.

2.
Cell ; 157(6): 1279-1291, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24906147

RESUMEN

Exercise training benefits many organ systems and offers protection against metabolic disorders such as obesity and diabetes. Using the recently identified isoform of PGC1-α (PGC1-α4) as a discovery tool, we report the identification of meteorin-like (Metrnl), a circulating factor that is induced in muscle after exercise and in adipose tissue upon cold exposure. Increasing circulating levels of Metrnl stimulates energy expenditure and improves glucose tolerance and the expression of genes associated with beige fat thermogenesis and anti-inflammatory cytokines. Metrnl stimulates an eosinophil-dependent increase in IL-4 expression and promotes alternative activation of adipose tissue macrophages, which are required for the increased expression of the thermogenic and anti-inflammatory gene programs in fat. Importantly, blocking Metrnl actions in vivo significantly attenuates chronic cold-exposure-induced alternative macrophage activation and thermogenic gene responses. Thus, Metrnl links host-adaptive responses to the regulation of energy homeostasis and tissue inflammation and has therapeutic potential for metabolic and inflammatory diseases.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Animales , Glucosa/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Factores de Crecimiento Nervioso/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Termogénesis , Factores de Transcripción/genética
3.
Nat Med ; 20(4): 408-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24658078

RESUMEN

Erythropoietin (EPO) stimulates proliferation of early-stage erythrocyte precursors and is widely used for the treatment of chronic anemia. However, several types of EPO-resistant anemia are characterized by defects in late-stage erythropoiesis, which is EPO independent. Here we investigated regulation of erythropoiesis using a ligand-trapping fusion protein (ACE-536) containing the extracellular domain of human activin receptor type IIB (ActRIIB) modified to reduce activin binding. ACE-536, or its mouse version RAP-536, produced rapid and robust increases in erythrocyte numbers in multiple species under basal conditions and reduced or prevented anemia in murine models. Unlike EPO, RAP-536 promoted maturation of late-stage erythroid precursors in vivo. Cotreatment with ACE-536 and EPO produced a synergistic erythropoietic response. ACE-536 bound growth differentiation factor-11 (GDF11) and potently inhibited GDF11-mediated Smad2/3 signaling. GDF11 inhibited erythroid maturation in mice in vivo and ex vivo. Expression of GDF11 and ActRIIB in erythroid precursors decreased progressively with maturation, suggesting an inhibitory role for GDF11 in late-stage erythroid differentiation. RAP-536 treatment also reduced Smad2/3 activation, anemia, erythroid hyperplasia and ineffective erythropoiesis in a mouse model of myelodysplastic syndromes (MDS). These findings implicate transforming growth factor-ß (TGF-ß) superfamily signaling in erythroid maturation and identify ACE-536 as a new potential treatment for anemia, including that caused by ineffective erythropoiesis.


Asunto(s)
Receptores de Activinas Tipo II , Anemia/sangre , Proteínas Morfogenéticas Óseas/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Factores de Diferenciación de Crecimiento/efectos de los fármacos , Hematínicos/farmacología , Síndromes Mielodisplásicos/sangre , Proteínas Recombinantes de Fusión/farmacología , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Quimioterapia Combinada , Recuento de Eritrocitos , Eritropoyetina/farmacología , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Haplorrinos , Humanos , Ligandos , Ratones , Ratas , Recuento de Reticulocitos , Transducción de Señal/efectos de los fármacos , Proteína Smad2/efectos de los fármacos , Proteína smad3/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 109(30): 12207-12, 2012 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-22761317

RESUMEN

Diseases such as osteoporosis are associated with reduced bone mass. Therapies to prevent bone loss exist, but there are few that stimulate bone formation and restore bone mass. Bone morphogenetic proteins (BMPs) are members of the TGFß superfamily, which act as pleiotropic regulators of skeletal organogenesis and bone homeostasis. Ablation of the BMPR1A receptor in osteoblasts increases bone mass, suggesting that inhibition of BMPR1A signaling may have therapeutic benefit. The aim of this study was to determine the skeletal effects of systemic administration of a soluble BMPR1A fusion protein (mBMPR1A-mFc) in vivo. mBMPR1A-mFc was shown to bind BMP2/4 specifically and with high affinity and prevent downstream signaling. mBMPR1A-mFc treatment of immature and mature mice increased bone mineral density, cortical thickness, trabecular bone volume, thickness and number, and decreased trabecular separation. The increase in bone mass was due to an early increase in osteoblast number and bone formation rate, mediated by a suppression of Dickkopf-1 expression. This was followed by a decrease in osteoclast number and eroded surface, which was associated with a decrease in receptor activator of NF-κB ligand (RANKL) production, an increase in osteoprotegerin expression, and a decrease in serum tartrate-resistant acid phosphatase (TRAP5b) concentration. mBMPR1A treatment also increased bone mass and strength in mice with bone loss due to estrogen deficiency. In conclusion, mBMPR1A-mFc stimulates osteoblastic bone formation and decreases bone resorption, which leads to an increase in bone mass, and offers a promising unique alternative for the treatment of bone-related disorders.


Asunto(s)
Enfermedades Óseas Metabólicas/prevención & control , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Huesos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Densidad Ósea/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Resorción Ósea/tratamiento farmacológico , Huesos/anatomía & histología , Huesos/fisiología , Cromatografía en Gel , Clonación Molecular , Densitometría , Electroforesis en Gel de Poliacrilamida , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Luciferasas , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteoprotegerina/metabolismo , Reacción en Cadena de la Polimerasa , Ligando RANK/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes de Fusión/administración & dosificación , Transducción de Señal/fisiología
5.
J Biol Chem ; 287(33): 27313-25, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22718755

RESUMEN

Activin receptor-like kinase 1 (ALK1), an endothelial cell-specific type I receptor of the TGF-ß superfamily, is an important regulator of normal blood vessel development as well as pathological tumor angiogenesis. As such, ALK1 is an important therapeutic target. Thus, several ALK1-directed agents are currently in clinical trials as anti-angiogenic cancer therapeutics. Given the biological and clinical importance of the ALK1 signaling pathway, we sought to elucidate the biophysical and structural basis underlying ALK1 signaling. The TGF-ß family ligands BMP9 and BMP10 as well as the three type II TGF-ß family receptors ActRIIA, ActRIIB, and BMPRII have been implicated in ALK1 signaling. Here, we provide a kinetic and thermodynamic analysis of BMP9 and BMP10 interactions with ALK1 and type II receptors. Our data show that BMP9 displays a significant discrimination in type II receptor binding, whereas BMP10 does not. We also report the crystal structure of a fully assembled ternary complex of BMP9 with the extracellular domains of ALK1 and ActRIIB. The structure reveals that the high specificity of ALK1 for BMP9/10 is determined by a novel orientation of ALK1 with respect to BMP9, which leads to a unique set of receptor-ligand interactions. In addition, the structure explains how BMP9 discriminates between low and high affinity type II receptors. Taken together, our findings provide structural and mechanistic insights into ALK1 signaling that could serve as a basis for novel anti-angiogenic therapies.


Asunto(s)
Receptores de Activinas Tipo II/química , Proteínas Morfogenéticas Óseas/química , Factores de Diferenciación de Crecimiento/química , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/uso terapéutico , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cristalografía por Rayos X , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
6.
Endocrinology ; 153(7): 3133-46, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22549226

RESUMEN

Obesity results from disproportionately high energy intake relative to energy expenditure. Many therapeutic strategies have focused on the intake side of the equation, including pharmaceutical targeting of appetite and digestion. An alternative approach is to increase energy expenditure through physical activity or adaptive thermogenesis. A pharmacological way to increase muscle mass and hence exercise capacity is through inhibition of the activin receptor type IIB (ActRIIB). Muscle mass and strength is regulated, at least in part, by growth factors that signal via ActRIIB. Administration of a soluble ActRIIB protein comprised of a form of the extracellular domain of ActRIIB fused to a human Fc (ActRIIB-Fc) results in a substantial muscle mass increase in normal mice. However, ActRIIB is also present on and mediates the action of growth factors in adipose tissue, although the function of this system is poorly understood. In the current study, we report the effect of ActRIIB-Fc to suppress diet-induced obesity and linked metabolic dysfunctions in mice fed a high-fat diet. ActRIIB-Fc induced a brown fat-like thermogenic gene program in epididymal white fat, as shown by robustly increased expression of the thermogenic genes uncoupling protein 1 and peroxisomal proliferator-activated receptor-γ coactivator 1α. Finally, we identified multiple ligands capable of reducing thermogenesis that represent likely target ligands for the ActRIIB-Fc effects on the white fat depots. These data demonstrate that novel therapeutic ActRIIB-Fc improves obesity and obesity-linked metabolic disease by both increasing skeletal muscle mass and by inducing a gene program of thermogenesis in the white adipose tissues.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Obesidad/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores Activados del Proliferador del Peroxisoma , Resonancia por Plasmón de Superficie , Termogénesis , Tomografía Computarizada por Rayos X/métodos , Factores de Transcripción
7.
Arterioscler Thromb Vasc Biol ; 32(3): 613-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22223731

RESUMEN

OBJECTIVE: The expression of bone morphogenetic proteins (BMPs) is enhanced in human atherosclerotic and calcific vascular lesions. Although genetic gain- and loss-of-function experiments in mice have supported a causal role of BMP signaling in atherosclerosis and vascular calcification, it remains uncertain whether BMP signaling might be targeted pharmacologically to ameliorate both of these processes. METHODS AND RESULTS: We tested the impact of pharmacological BMP inhibition on atherosclerosis and calcification in LDL receptor-deficient (LDLR-/-) mice. LDLR-/- mice fed a high-fat diet developed abundant vascular calcification within 20 weeks. Prolonged treatment of LDLR-/- mice with the small molecule BMP inhibitor LDN-193189 was well-tolerated and potently inhibited development of atheroma, as well as associated vascular inflammation, osteogenic activity, and calcification. Administration of recombinant BMP antagonist ALK3-Fc replicated the antiatherosclerotic and anti-inflammatory effects of LDN-193189. Treatment of human aortic endothelial cells with LDN-193189 or ALK3-Fc abrogated the production of reactive oxygen species induced by oxidized LDL, a known early event in atherogenesis. Unexpectedly, treatment of mice with LDN-193189 lowered LDL serum cholesterol by 35% and markedly decreased hepatosteatosis without inhibiting HMG-CoA reductase activity. Treatment with BMP2 increased, whereas LDN-193189 or ALK3-Fc inhibited apolipoprotein B100 secretion in HepG2 cells, suggesting that BMP signaling contributes to the regulation of cholesterol biosynthesis. CONCLUSION: These results definitively implicate BMP signaling in atherosclerosis and calcification, while uncovering a previously unidentified role for BMP signaling in LDL cholesterol metabolism. BMP inhibition may be helpful in the treatment of atherosclerosis and associated vascular calcification.


Asunto(s)
Aterosclerosis/prevención & control , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Fármacos Cardiovasculares/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/prevención & control , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , LDL-Colesterol/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Femenino , Células Hep G2 , Humanos , Lipoproteínas LDL/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Calcificación Vascular/etiología , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
8.
J Biol Chem ; 286(34): 30034-46, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21737454

RESUMEN

Endoglin (CD105), a transmembrane protein of the transforming growth factor ß superfamily, plays a crucial role in angiogenesis. Mutations in endoglin result in the vascular defect known as hereditary hemorrhagic telangiectasia (HHT1). The soluble form of endoglin was suggested to contribute to the pathogenesis of preeclampsia. To obtain further insight into its function, we cloned, expressed, purified, and characterized the extracellular domain (ECD) of mouse and human endoglin fused to an immunoglobulin Fc domain. We found that mouse and human endoglin ECD-Fc bound directly, specifically, and with high affinity to bone morphogenetic proteins 9 and 10 (BMP9 and BMP10) in surface plasmon resonance (Biacore) and cell-based assays. We performed a function mapping analysis of the different domains of endoglin by examining their contributions to the selectivity and biological activity of the protein. The BMP9/BMP10 binding site was localized to the orphan domain of human endoglin composed of the amino acid sequence 26-359. We established that endoglin and type II receptors bind to overlapping sites on BMP9. In the in vivo chick chorioallantoic membrane assay, the mouse and the truncated human endoglin ECD-Fc both significantly reduced VEGF-induced vessel formation. Finally, murine endoglin ECD-Fc acted as an anti-angiogenic factor that decreased blood vessel sprouting in VEGF/FGF-induced angiogenesis in in vivo angioreactors and reduced the tumor burden in the colon-26 mouse tumor model. Together our findings indicate an important role of soluble endoglin ECD in the regulation of angiogenesis and highlight efficacy of endoglin-Fc as a potential anti-angiogenesis therapeutic agent.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antígenos CD/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Factor 2 de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intracelular/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Sitios de Unión , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Endoglina , Factor 2 de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
9.
Muscle Nerve ; 43(5): 694-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21462203

RESUMEN

INTRODUCTION: In this study we investigated the action of RAP-031, a soluble activin receptor type IIB (ActRIIB) comprised of a form of the ActRIIB extracellular domain linked to a murine Fc, and the NF-κB inhibitor, ursodeoxycholic acid (UDCA), on the whole body strength of mdx mice. METHODS: The whole body tension (WBT) method of assessing the forward pulling tension (FPT) exerted by dystrophic (mdx) mice was used. RESULTS: RAP-031 produced a 41% increase in body mass and a 42.5% increase in FPT without altering the FPT normalized for body mass (WBT). Coadministration of RAP-031 with UDCA produced increases in FPT that were associated with an increase in WBT. CONCLUSIONS: Myostatin inhibition increases muscle mass without altering the fundamental weakness characteristic of dystrophic muscle. Cotreatment with an NF-κB inhibitor potentiates the effects of myostatin inhibition in improving FPT in mdx mice.


Asunto(s)
Receptores de Activinas Tipo II/fisiología , Tono Muscular/fisiología , Músculo Esquelético/fisiología , Distrofia Muscular Animal/fisiopatología , Receptores de Activinas Tipo II/farmacología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Tono Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/genética , Solubilidad
10.
Am J Pathol ; 178(3): 1287-97, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356379

RESUMEN

The activin receptor type IIB (ActRIIB) is a transmembrane receptor for transforming growth factor-ß superfamily members, including myostatin, that are involved in the negative regulation of skeletal muscle mass. We tested the translational hypothesis that blocking ligand binding to ActRIIB for 12 weeks would stimulate skeletal muscle growth and improve muscle function in the mdx mouse. ActRIIB was targeted using a novel inhibitor comprised of the extracellular portion of the ActRIIB fused to the Fc portion of murine IgG (sActRIIB), at concentrations of 1.0 and 10.0 mg/kg(-1) body weight. After 12 weeks of treatment, the 10.0 mg/kg(-1) dose caused a 27% increase in body weight with a concomitant 33% increase in lean muscle mass. Absolute force production of the extensor digitorum longus muscle ex vivo was higher in mice after treatment with either dose of sActRIIB, and the specific force was significantly higher after the lower dose (1.0 mg/kg(-1)), indicating functional improvement in the muscle. Circulating creatine kinase levels were significantly lower in mice treated with sActRIIB, compared with control mice. These data show that targeting the ActRIIB improves skeletal muscle mass and functional strength in the mdx mouse model of DMD, providing a therapeutic rationale for use of this molecule in treating skeletal myopathies.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Recuperación de la Función/fisiología , Receptores de Activinas Tipo II/metabolismo , Animales , Fenómenos Biomecánicos , Peso Corporal , Creatina Quinasa/sangre , Hidroxiprolina/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular/fisiología , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/patología , Tamaño de los Órganos
11.
Blood ; 117(18): 4915-23, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21393479

RESUMEN

Anemia of inflammation develops in settings of chronic inflammatory, infectious, or neoplastic disease. In this highly prevalent form of anemia, inflammatory cytokines, including IL-6, stimulate hepatic expression of hepcidin, which negatively regulates iron bioavailability by inactivating ferroportin. Hepcidin is transcriptionally regulated by IL-6 and bone morphogenetic protein (BMP) signaling. We hypothesized that inhibiting BMP signaling can reduce hepcidin expression and ameliorate hypoferremia and anemia associated with inflammation. In human hepatoma cells, IL-6-induced hepcidin expression, an effect that was inhibited by treatment with a BMP type I receptor inhibitor, LDN-193189, or BMP ligand antagonists noggin and ALK3-Fc. In zebrafish, the induction of hepcidin expression by transgenic expression of IL-6 was also reduced by LDN-193189. In mice, treatment with IL-6 or turpentine increased hepcidin expression and reduced serum iron, effects that were inhibited by LDN-193189 or ALK3-Fc. Chronic turpentine treatment led to microcytic anemia, which was prevented by concurrent administration of LDN-193189 or attenuated when LDN-193189 was administered after anemia was established. Our studies support the concept that BMP and IL-6 act together to regulate iron homeostasis and suggest that inhibition of BMP signaling may be an effective strategy for the treatment of anemia of inflammation.


Asunto(s)
Anemia/etiología , Anemia/prevención & control , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Inflamación/complicaciones , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/antagonistas & inhibidores , Proteínas Portadoras/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Hep G2 , Hepcidinas , Humanos , Interleucina-6/farmacología , Ratones , Ratones Endogámicos C57BL , Pirazoles/farmacología , Pirimidinas/farmacología , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Trementina/toxicidad , Pez Cebra , Proteínas de Pez Cebra/metabolismo
12.
Am J Pathol ; 178(2): 784-93, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21281811

RESUMEN

X-linked myotubular myopathy (XLMTM) is a congenital disorder caused by deficiency of the lipid phosphatase, myotubularin. Patients with XLMTM often have severe perinatal weakness that requires mechanical ventilation to prevent death from respiratory failure. Muscle biopsy specimens from patients with XLMTM exhibit small myofibers with central nuclei and central aggregations of organelles in many cells. It was postulated that therapeutically increasing muscle fiber size would cause symptomatic improvement in myotubularin deficiency. Recent studies have elucidated an important role for the activin-receptor type IIB (ActRIIB) in regulation of muscle growth and have demonstrated that ActRIIB inhibition results in significant muscle hypertrophy. To evaluate whether promoting muscle hypertrophy can attenuate symptoms resulting from myotubularin deficiency, the effect of ActRIIB-mFC treatment was determined in myotubularin-deficient (Mtm1δ4) mice. Compared with wild-type mice, untreated Mtm1δ4 mice have decreased body weight, skeletal muscle hypotrophy, and reduced survival. Treatment of Mtm1δ4 mice with ActRIIB-mFC produced a 17% extension of lifespan, with transient increases in weight, forelimb grip strength, and myofiber size. Pathologic analysis of Mtm1δ4 mice during treatment revealed that ActRIIB-mFC produced marked hypertrophy restricted to type 2b myofibers, which suggests that oxidative fibers in Mtm1δ4 animals are incapable of a hypertrophic response in this setting. These results support ActRIIB-mFC as an effective treatment for the weakness observed in myotubularin deficiency.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Longevidad/fisiología , Fuerza Muscular/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Receptores de Activinas Tipo II/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Miembro Anterior/efectos de los fármacos , Miembro Anterior/fisiología , Gravitación , Fuerza de la Mano/fisiología , Longevidad/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Miostatina/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Análisis de Supervivencia
13.
J Bone Miner Res ; 25(12): 2633-46, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20533325

RESUMEN

Cancers that grow in bone, such as myeloma and breast cancer metastases, cause devastating osteolytic bone destruction. These cancers hijack bone remodeling by stimulating osteoclastic bone resorption and suppressing bone formation. Currently, treatment is targeted primarily at blocking bone resorption, but this approach has achieved only limited success. Stimulating osteoblastic bone formation to promote repair is a novel alternative approach. We show that a soluble activin receptor type IIA fusion protein (ActRIIA.muFc) stimulates osteoblastogenesis (p < .01), promotes bone formation (p < .01) and increases bone mass in vivo (p < .001). We show that the development of osteolytic bone lesions in mice bearing murine myeloma cells is caused by both increased resorption (p < .05) and suppression of bone formation (p < .01). ActRIIA.muFc treatment stimulates osteoblastogenesis (p < .01), prevents myeloma-induced suppression of bone formation (p < .05), blocks the development of osteolytic bone lesions (p < .05), and increases survival (p < .05). We also show, in a murine model of breast cancer bone metastasis, that ActRIIA.muFc again prevents bone destruction (p < .001) and inhibits bone metastases (p < .05). These findings show that stimulating osteoblastic bone formation with ActRIIA.muFc blocks the formation of osteolytic bone lesions and bone metastases in models of myeloma and breast cancer and paves the way for new approaches to treating this debilitating aspect of cancer.


Asunto(s)
Activinas/metabolismo , Neoplasias Óseas/complicaciones , Resorción Ósea/etiología , Resorción Ósea/prevención & control , Osteogénesis , Transducción de Señal , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/fisiopatología , Neoplasias Óseas/secundario , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Calcificación Fisiológica/efectos de los fármacos , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Mieloma Múltiple/complicaciones , Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Trasplante de Neoplasias , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteogénesis/efectos de los fármacos , Osteólisis/sangre , Osteólisis/complicaciones , Osteólisis/fisiopatología , Osteólisis/prevención & control , Paraproteínas/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos
14.
Endocrinology ; 151(9): 4289-300, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20573726

RESUMEN

Androgen deprivation, a consequence of hypogonadism, certain cancer treatments, or normal aging in men, leads to loss of muscle mass, increased adiposity, and osteoporosis. In the present study, using a soluble chimeric form of activin receptor type IIB (ActRIIB) we sought to offset the adverse effects of androgen deprivation on muscle, adipose tissue, and bone. Castrated (ORX) or sham-operated (SHAM) mice received either TBS [vehicle-treated (VEH)] or systemic administration of ActRIIB-mFc, a soluble fusion protein comprised of a form of the extracellular domain of ActRIIB fused to a murine IgG2aFc subunit. In vivo body composition imaging demonstrated that ActRIIB-mFc treatment results in increased lean tissue mass of 23% in SHAM mice [19.02 +/- 0.42 g (VEH) versus 23.43 +/- 0.35 g (ActRIIB-mFc), P < 0.00001] and 26% in ORX mice [15.59 +/- 0.26 g (VEH) versus 19.78 +/- 0.26 g (ActRIIB-mFc), P < 0.00001]. Treatment also caused a decrease in adiposity of 30% in SHAM mice [5.03 +/- 0.48 g (VEH) versus 3.53 +/- 0.19 g (ActRIIB-mFc), NS] and 36% in ORX mice [7.12 +/- 0.53 g (VEH) versus 4.57 +/- 0.28 g (ActRIIB-mFc), P < 0.001]. These changes were also accompanied by altered serum levels of leptin, adiponectin, and insulin, as well as by prevention of steatosis (fatty liver) in ActRIIB-mFc-treated ORX mice. Finally, ActRIIB-mFc prevented loss of bone mass in ORX mice as assessed by whole body dual x-ray absorptiometry and micro-computed tomography of proximal tibias. The data demonstrate that treatment with ActRIIB-mFc restored muscle mass, adiposity, and bone quality to normal levels in a mouse model of androgen deprivation, thereby alleviating multiple adverse consequences of such therapy.


Asunto(s)
Receptores de Activinas Tipo II/farmacología , Antagonistas de Andrógenos/farmacología , Composición Corporal/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Receptores de Activinas Tipo II/genética , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Análisis de Varianza , Animales , Peso Corporal/efectos de los fármacos , Línea Celular , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/sangre , Obesidad/prevención & control , Orquiectomía , Distribución Aleatoria , Proteínas Recombinantes de Fusión/farmacología , Solubilidad
15.
J Biol Chem ; 285(27): 21037-48, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20385559

RESUMEN

The single transmembrane domain serine/threonine kinase activin receptor type IIB (ActRIIB) has been proposed to bind key regulators of skeletal muscle mass development, including the ligands GDF-8 (myostatin) and GDF-11 (BMP-11). Here we provide a detailed kinetic characterization of ActRIIB binding to several low and high affinity ligands using a soluble activin receptor type IIB-Fc chimera (ActRIIB.Fc). We show that both GDF-8 and GDF-11 bind the extracellular domain of ActRIIB with affinities comparable with those of activin A, a known high affinity ActRIIB ligand, whereas BMP-2 and BMP-7 affinities for ActRIIB are at least 100-fold lower. Using site-directed mutagenesis, we demonstrate that ActRIIB binds GDF-11 and activin A in different ways such as, for example, substitutions in ActRIIB Leu(79) effectively abolish ActRIIB binding to activin A yet not to GDF-11. Native ActRIIB has four isoforms that differ in the length of the C-terminal portion of their extracellular domains. We demonstrate that the C terminus of the ActRIIB extracellular domain is crucial for maintaining biological activity of the ActRIIB.Fc receptor chimera. In addition, we show that glycosylation of ActRIIB is not required for binding to activin A or GDF-11. Together, our findings reveal binding specificity and activity determinants of the ActRIIB receptor that combine to effect specificity in the activation of distinct signaling pathways.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , ADN Complementario/genética , Genes Reporteros , Humanos , Ligandos , Mutagénesis , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/metabolismo , Miostatina/química , Miostatina/metabolismo , Plásmidos/genética , Activadores Plasminogénicos/química , Activadores Plasminogénicos/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(11): 5124-9, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20194748

RESUMEN

Understanding the pathogenesis of cancer-related bone disease is crucial to the discovery of new therapies. Here we identify activin A, a TGF-beta family member, as a therapeutically amenable target exploited by multiple myeloma (MM) to alter its microenvironmental niche favoring osteolysis. Increased bone marrow plasma activin A levels were found in MM patients with osteolytic disease. MM cell engagement of marrow stromal cells enhanced activin A secretion via adhesion-mediated JNK activation. Activin A, in turn, inhibited osteoblast differentiation via SMAD2-dependent distal-less homeobox-5 down-regulation. Targeting activin A by a soluble decoy receptor reversed osteoblast inhibition, ameliorated MM bone disease, and inhibited tumor growth in an in vivo humanized MM model, setting the stage for testing in human clinical trials.


Asunto(s)
Activinas/metabolismo , Mieloma Múltiple/complicaciones , Osteólisis/etiología , Activinas/antagonistas & inhibidores , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Osteoblastos/patología , Osteólisis/patología , Receptores de Superficie Celular/metabolismo , Proteína Smad2/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
17.
Mol Cancer Ther ; 9(2): 379-88, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20124460

RESUMEN

Activin receptor-like kinase-1 (ALK1) is a type I, endothelial cell-specific member of the transforming growth factor-beta superfamily of receptors known to play an essential role in modulating angiogenesis and vessel maintenance. In the present study, we sought to examine the angiogenic and tumorigenic effects mediated upon the inhibition of ALK1 signaling using a soluble chimeric protein (ALK1-Fc). Of 29 transforming growth factor-beta-related ligands screened by surface plasmon resonance, only bone morphogenetic protein (BMP9) and BMP10 displayed high-affinity binding to ALK1-Fc. In cell-based assays, ALK1-Fc inhibited BMP9-mediated Id-1 expression in human umbilical vein endothelial cells and inhibited cord formation by these cells on a Matrigel substrate. In a chick chorioallantoic membrane assay, ALK1-Fc reduced vascular endothelial growth factor-, fibroblast growth factor-, and BMP10-mediated vessel formation. The growth of B16 melanoma explants was also inhibited significantly by ALK1-Fc in this assay. Finally, ALK1-Fc treatment reduced tumor burden in mice receiving orthotopic grafts of MCF7 mammary adenocarcinoma cells. These data show the efficacy of chimeric ALK1-Fc proteins in mitigating vessel formation and support the view that ALK1-Fc is a powerful antiangiogenic agent capable of blocking vascularization.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Células CHO , Cricetinae , Cricetulus , Células Endoteliales/citología , Endotelio Vascular/citología , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Resonancia por Plasmón de Superficie , Telangiectasia Hemorrágica Hereditaria/metabolismo
18.
Bone ; 46(4): 1082-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20080223

RESUMEN

Activin A belongs to the TGF-beta superfamily and plays an important role in bone metabolism. It was reported that a soluble form of extracellular domain of the activin receptor type IIA (ActRIIA) fused to the Fc domain of murine IgG, an activin antagonist, has an anabolic effect on bone in intact and ovariectomized mice. The present study was designed to examine the skeletal effect of human ActRIIA-IgG1-Fc (ACE-011) in non-human primates. Young adult female Cynomolgus monkeys were given a biweekly subcutaneous injection of either 10mg/kg ACE-011 or vehicle (VEH) for 3months. Treatment effects were evaluated by histomorphometric analysis of the distal femur, femoral midshaft, femoral neck and 12th thoracic vertebrae, by muCT analysis of femoral neck and by biomarkers of bone turnover. Compared to VEH, at the distal femur ACE-011-treated monkeys had significantly increased cancellous bone volume (+93%), bone formation rate per bone surface (+166%) and osteoblast surface (+196%) indicating an anabolic action. Monkeys treated with ACE-011 also had decreased osteoclast surface and number. No differences were observed in parameters of cortical bone at the midshaft of the femur. Similar to distal femur, ACE-011-treated monkeys had significantly greater cancellous bone volume, bone formation rate and osteoblast surface at the femoral neck relative to VEH. A significant increase in bone formation rate and osteoblast surface with a decrease in osteoclast surface was observed in thoracic vertebrae. muCT analysis of femoral neck indicated more plate-like structure in ACE-011-treated monkeys. Monkeys treated with ACE-011 had no effect on serum bone-specific alkaline phosphatase and CTX at the end of the study. These observations demonstrate that ACE-011 is a dual anabolic-antiresorptive compound, improving cancellous bone volume by promoting bone formation and inhibiting bone resorption in non-human primates. Thus, soluble ActRIIA fusion protein may be useful in the prevention and/or treatment of osteoporosis and other diseases involving accelerated bone loss.


Asunto(s)
Activinas/metabolismo , Densidad Ósea/fisiología , Fémur/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Vértebras Torácicas/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Recuento de Células , Colágeno Tipo I/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Fémur/efectos de los fármacos , Macaca fascicularis , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Distribución Aleatoria , Proteínas Recombinantes de Fusión/metabolismo , Estadísticas no Paramétricas , Vértebras Torácicas/efectos de los fármacos
19.
J Exp Med ; 207(1): 85-100, 2010 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-20065063

RESUMEN

Members of the transforming growth factor beta (TGF-beta) family have been genetically linked to vascular formation during embryogenesis. However, contradictory studies about the role of TGF-beta and other family members with reported vascular functions, such as bone morphogenetic protein (BMP) 9, in physiological and pathological angiogenesis make the need for mechanistic studies apparent. We demonstrate, by genetic and pharmacological means, that the TGF-beta and BMP9 receptor activin receptor-like kinase (ALK) 1 represents a new therapeutic target for tumor angiogenesis. Diminution of ALK1 gene dosage or systemic treatment with the ALK1-Fc fusion protein RAP-041 retarded tumor growth and progression by inhibition of angiogenesis in a transgenic mouse model of multistep tumorigenesis. Furthermore, RAP-041 significantly impaired the in vitro and in vivo angiogenic response toward vascular endothelial growth factor A and basic fibroblast growth factor. In seeking the mechanism for the observed effects, we uncovered an unexpected signaling synergy between TGF-beta and BMP9, through which the combined action of the two factors augmented the endothelial cell response to angiogenic stimuli. We delineate a decisive role for signaling by TGF-beta family members in tumor angiogenesis and offer mechanistic insight for the forthcoming clinical development of drugs blocking ALK1 in oncology.


Asunto(s)
Receptores de Activinas Tipo II , Receptores de Activinas Tipo I , Células Endoteliales/metabolismo , Fragmentos Fc de Inmunoglobulinas/farmacología , Neoplasias Experimentales , Neovascularización Patológica , Proteínas Recombinantes de Fusión/farmacología , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo I/farmacología , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/farmacología , Animales , Línea Celular , Células Endoteliales/patología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Dosificación de Gen/genética , Factor 2 de Diferenciación de Crecimiento/genética , Factor 2 de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Am J Physiol Regul Integr Comp Physiol ; 298(1): R96-R103, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19864340

RESUMEN

Hypoxia, or reduced oxygen, occurs in a variety of clinical and environmental situations. Hypoxic exposure is associated with decreased muscle mass and a concomitant reduction in exercise capacity, although the exact mechanisms are not completely understood. The activin type IIB receptor (ActRIIB) is a receptor for transforming growth factor-beta (TGFbeta) superfamily members that are involved in the negative regulation of lean tissue mass. Given that hypoxia has negative effects on muscle mass and function and that modulation of the ActRIIB has been shown to increase muscle mass, we tested the hypothesis that pharmacological targeting of the ActRIIB for 2 wk would attenuate the loss of muscle mass and function in mice after exposure to normobaric hypoxia. ActRIIB modulation was achieved using a soluble activin receptor/Fc fusion protein (sActRIIB) in mice housed in a hypoxic chamber for 1 or 2 wk. Hypoxia induced a reduction in body weight in PBS- and sActRIIB-treated mice, although sActRIIB-treated mice remained larger throughout the hypoxic exposure. The absolute forces generated by extensor digitorum longus muscles were also significantly greater in sActRIIB- than PBS-treated mice and were more resistant to eccentric contraction-induced force drop after eccentric lengthening contractions. In summary, sActRIIB pretreatment attenuated hypoxia-induced muscle dysfunction. These data suggest that targeting the ActRIIB is an effective strategy to counter hypoxia-induced muscle dysfunction and to preacclimatize to hypoxia in clinical or high-altitude settings.


Asunto(s)
Receptores de Activinas Tipo II/farmacología , Hipoxia/fisiopatología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Esquelético/patología , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA