Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Calcif Tissue Int ; 99(5): 489-499, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27395059

RESUMEN

Abaloparatide is a novel, potent and selective activator of parathyroid hormone receptor 1 (PTHR1) under clinical development for the treatment of osteoporosis. We assessed the effect of 6 weeks of abaloparatide on bone mass, microarchitecture, quality and strength in ovariectomized (OVX) rats. After 8 weeks of post-surgical bone depletion (baseline), OVX rats (n = 20-21/group) received daily subcutaneous vehicle (OVX-Veh) or abaloparatide at 5 or 20 µg/kg. Sham-operated control rats (n = 24) received vehicle. Areal bone mineral density (aBMD) of the lumbar spine (L4), total femur and femur diaphysis was measured at baseline and after 6 weeks of treatment. Femur and vertebral bone architecture and mechanical properties were assessed at the end of the treatment phase. At baseline, OVX-Veh rats exhibited significantly lower aBMD relative to Sham controls. Treatment of OVX rats with abaloparatide at 5 or 20 µg/kg/day increased aBMD dose-dependently in the lumbar spine, total femur and femur diaphysis to levels exceeding OVX-Veh or Sham controls. The abaloparatide 5 and 20 µg/kg groups had improved trabecular microarchitecture relative to OVX vehicle, with trabecular BV/TV exceeding OVX-Veh control values by 57 and 78 % (respectively) at the lumbar spine, and by 145 and 270 % at the distal femur. Femur diaphyseal cortical volume and thickness were significantly greater in the abaloparatide 20 µg/kg group relative to OVX vehicle or Sham controls. Bone strength parameters of the femur diaphysis, femur neck and L4 vertebra were significantly improved in the OVX-ABL groups relative to OVX-Veh controls. Bone mass-strength relationships and estimated intrinsic strength properties suggested maintained or improved bone quality with abaloparatide. These data demonstrate skeletal restoration via abaloparatide treatment of osteopenic OVX rats, in association with improved trabecular microarchitecture, cortical geometry and bone strength at sites that have clinical relevance in patients with osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/prevención & control , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Absorciometría de Fotón , Animales , Femenino , Fémur/efectos de los fármacos , Vértebras Lumbares/efectos de los fármacos , Ovariectomía , Ratas , Ratas Sprague-Dawley , Microtomografía por Rayos X
2.
Arterioscler Thromb Vasc Biol ; 35(4): 911-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25614286

RESUMEN

OBJECTIVE: Studies of mice with mild Marfan syndrome (MFS) have correlated the development of thoracic aortic aneurysm (TAA) with improper stimulation of noncanonical (Erk-mediated) TGFß signaling by the angiotensin type I receptor (AT1r). This correlation was largely based on comparable TAA modifications by either systemic TGFß neutralization or AT1r antagonism. However, subsequent investigations have called into question some key aspects of this mechanism of arterial disease in MFS. To resolve these controversial points, here we made a head-to-head comparison of the therapeutic benefits of TGFß neutralization and AT1r antagonism in mice with progressively severe MFS (Fbn1(mgR/mgR) mice). APPROACH AND RESULTS: Aneurysm growth, media degeneration, aortic levels of phosphorylated Erk and Smad proteins and the average survival of Fbn1(mgR/mgR) mice were compared after a ≈3-month-long treatment with placebo and either the AT1r antagonist losartan or the TGFß-neutralizing antibody 1D11. In contrast to the beneficial effect of losartan, TGFß neutralization either exacerbated or mitigated TAA formation depending on whether treatment was initiated before (postnatal day 16; P16) or after (P45) aneurysm formation, respectively. Biochemical evidence-related aneurysm growth with Erk-mediated AT1r signaling, and medial degeneration with TGFß hyperactivity that was in part AT1r dependent. Importantly, P16-initiated treatment with losartan combined with P45-initiated administration of 1D11 prevented death of Fbn1(mgR/mgR) mice from ruptured TAA. CONCLUSIONS: By demonstrating that promiscuous AT1r and TGFß drive partially overlapping processes of arterial disease in MFS mice, our study argues for a therapeutic strategy against TAA that targets both signaling pathways although sparing the early protective role of TGFß.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Anticuerpos Neutralizantes/farmacología , Aorta Torácica/efectos de los fármacos , Aneurisma de la Aorta Torácica/prevención & control , Losartán/farmacología , Síndrome de Marfan/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/patología , Rotura de la Aorta/genética , Rotura de la Aorta/metabolismo , Rotura de la Aorta/patología , Rotura de la Aorta/prevención & control , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas de Microfilamentos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Fosforilación , Receptor de Angiotensina Tipo 1/metabolismo , Proteína Smad2/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
3.
Am J Pathol ; 178(6): 2611-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641384

RESUMEN

Respiratory function is the main cause of mortality in patients with Duchenne muscular dystrophy (DMD). Elevated levels of TGF-ß play a key role in the pathophysiology of DMD. To determine whether therapeutic attenuation of TGF-ß signaling improves respiratory function, mdx mice were treated from 2 weeks of age to 2 months or 9 months of age with either 1D11 (a neutralizing antibody to all three isoforms of TGF-ß), losartan (an angiotensin receptor antagonist), or a combination of the two agents. Respiratory function was measured in nonanesthetized mice by plethysmography. The 9-month-old mdx mice had elevated Penh values and decreased breathing frequency, due primarily to decreased inspiratory flow rate. All treatments normalized Penh values and increased peak inspiratory flow, leading to decreased inspiration times and breathing frequency. Additionally, forelimb grip strength was improved after 1D11 treatment at both 2 and 9 months of age, whereas, losartan improved grip strength only at 2 months. Decreased serum creatine kinase levels (significant improvement for all groups), increased diaphragm muscle fiber density, and decreased hydroxyproline levels (significant improvement for 1D11 only) also suggested improved muscle function after treatment. For all endpoints, 1D11 was equivalent or superior to losartan; coadministration of the two agents was not superior to 1D11 alone. In conclusion, TGF-ß antagonism may be a useful therapeutic approach for treating DMD patients.


Asunto(s)
Respiración , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Creatina Quinasa/sangre , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Diafragma/patología , Diafragma/fisiopatología , Relación Dosis-Respuesta a Droga , Enalapril/administración & dosificación , Enalapril/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Fuerza de la Mano/fisiología , Hidroxiprolina/metabolismo , Inflamación/sangre , Inflamación/metabolismo , Inflamación/patología , Losartán/administración & dosificación , Losartán/farmacología , Ratones , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Miogenina/metabolismo , Tamaño de los Órganos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Respiración/efectos de los fármacos , Pruebas de Función Respiratoria , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
4.
J Med Chem ; 50(18): 4295-303, 2007 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-17691760

RESUMEN

The early and later eluting [(99m)TcO]depreotide products on RP-HPLC were confirmed to be the anti and syn diastereomers, respectively, based on proton NMR and circular dichroism spectroscopy. NMR provided evidence of a folded, conformationally constrained structure for the syn diastereomer. The syn diastereomer is predominant (anti/syn approximately 10:90) in the [(99m)TcO]depreotide preparation and shows a slightly higher affinity (IC50 = 0.15 nM) for the somatostatin receptor than the anti diastereomer (IC50 = 0.89 nM). Both diastereomers showed higher binding affinities than the free peptide (IC(50) = 7.4 nM). Biodistribution studies in AR42J tumor xenograft nude mice also showed higher tumor uptake for syn [(99m)TcO]depreotide (6.58% ID/g) than for the anti [(99m)TcO]depreotide (3.38% ID/g). Despite the differences in biological efficacy, the favorable binding affinity, tumor uptake, and tumor-to-background ratio results for both diastereomeric species predict that both are effective for imaging somatostatin receptor-positive tumors in vivo.


Asunto(s)
Neoplasias/diagnóstico por imagen , Compuestos de Organotecnecio/aislamiento & purificación , Radiofármacos/aislamiento & purificación , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Animales , Línea Celular Tumoral , Dicroismo Circular , Femenino , Marcaje Isotópico , Espectroscopía de Resonancia Magnética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias/metabolismo , Compuestos de Organotecnecio/química , Compuestos de Organotecnecio/farmacocinética , Neoplasias Pancreáticas , Ensayo de Unión Radioligante , Cintigrafía , Radiofármacos/química , Radiofármacos/farmacocinética , Ratas , Somatostatina/química , Somatostatina/aislamiento & purificación , Somatostatina/farmacocinética , Estereoisomerismo , Distribución Tisular
5.
J Med Chem ; 50(6): 1354-64, 2007 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-17315859

RESUMEN

Somatostatin derivative peptides previously designed for radiodiagnostic purposes (99mTc P829 or 99mTc depreotide) were reoptimized for radiotherapy of tumors with rhenium radioisotopes. An optimized pharmacophore peptide P1839 was derived by in vitro binding affinity assay to AR42J rat pancreatic tumor cell membranes. Peptides with chelating domains and their oxorhenium(V) complexes were tested in vitro for binding to NCI H69 human SCLC tumor membranes. Further optimization entailed radiolabeling with 99mTc and biodistribution in an AR42J xenograft mouse model. Kidney uptake was decreased substantially by removing positively charged residues. Neutral N3S diamide amine thiol chelators with no adjacent positive charges had the best overall properties. Substituting an aromatic amino acid into the chelator approximately doubled the tumor uptake. The final optimized peptide P2045 (39) radiolabeled with 99mTc exhibited increased tumor uptake ( approximately 25 %ID/g at 1.5 h), lower kidney uptake ( approximately 4.8 %ID/g at 1.5 h), and extensive urinary excretion (59 %ID at 1.5 h). Finally, comparison biodistribution studies between 99mTc and 188Re (39) showed a good correlation between the two metal complexes and demonstrated prolonged tumor retention (> or =24 h).


Asunto(s)
Quelantes/síntesis química , Compuestos Organometálicos/síntesis química , Péptidos/síntesis química , Radioisótopos , Radiofármacos/síntesis química , Receptores de Somatostatina/metabolismo , Renio , Animales , Línea Celular Tumoral , Quelantes/química , Femenino , Humanos , Marcaje Isotópico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacocinética , Péptidos/química , Ensayo de Unión Radioligante , Radiofármacos/química , Radiofármacos/farmacocinética , Ratas , Relación Estructura-Actividad , Tecnecio , Distribución Tisular , Trasplante Heterólogo
6.
J Nucl Med ; 55(12): 2020-5, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25359879

RESUMEN

UNLABELLED: P2045 is a peptide analog of somatostatin with picomolar affinity for the somatostatin receptor subtype 2 (SSTR2) upregulated in some pancreatic tumors. Studies were conducted in rat AR42J pancreatic tumor xenograft mice to determine whether (188)Re-P2045 could inhibit the growth of pancreatic cancer in an animal model. METHODS: (188)Re-P2045 was intravenously administered every 3 d for 16 d to nude mice with AR42J tumor xenografts that were approximately 20 mm(3) at study initiation. Tumor volumes were recorded throughout the dosing period. At necropsy, all tissues were assessed for levels of radioactivity and evaluated for histologic abnormalities. Clinical chemistry and hematology parameters were determined from terminal blood samples. The affinity of nonradioactive (185/187)Re-P2045 for somatostatin receptors was compared in human NCI-H69 and rat AR42J tumor cell membranes expressing predominantly SSTR2. RESULTS: In the 1.85- and 5.55-MBq groups, tumor growth was inhibited in a dose-dependent fashion. In the 11.1-MBq group, tumor growth was completely inhibited throughout the dosing period and for 12 d after the last administered dose. The radioactivity level in tumors 4 h after injection was 10 percentage injected dose per gram, which was 2-fold higher than in the kidneys. (188)Re-P2045 was well tolerated in all dose groups, with no adverse clinical, histologic, or hematologic findings. The nonradioactive (185/187)Re-P2045 bound more avidly (0.2 nM) to SSTR2 in human than rat tumor membranes, suggesting that these studies are relevant to human studies. CONCLUSION: (188)Re-P2045 is a promising therapeutic candidate for patients with somatostatin receptor-positive cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Complejos de Coordinación/uso terapéutico , Antagonistas de Hormonas/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Péptidos/uso terapéutico , Radiofármacos/uso terapéutico , Somatostatina/análogos & derivados , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Complejos de Coordinación/efectos adversos , Complejos de Coordinación/farmacocinética , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Péptidos/efectos adversos , Péptidos/farmacocinética , Radiofármacos/efectos adversos , Radiofármacos/farmacocinética , Ratas , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Ther Nucleic Acids ; 3: e206, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25350581

RESUMEN

Pompe disease is an autosomal recessive disorder caused by a deficiency of acid α-glucosidase (GAA; EC 3.2.1.20) and the resultant progressive lysosomal accumulation of glycogen in skeletal and cardiac muscles. Enzyme replacement therapy using recombinant human GAA (rhGAA) has proven beneficial in addressing several aspects of the disease such as cardiomyopathy and aberrant motor function. However, residual muscle weakness, hearing loss, and the risks of arrhythmias and osteopenia persist despite enzyme therapy. Here, we evaluated the relative merits of substrate reduction therapy (by inhibiting glycogen synthesis) as a potential adjuvant strategy. A phosphorodiamidate morpholino oligonucleotide (PMO) designed to invoke exon skipping and premature stop codon usage in the transcript for muscle specific glycogen synthase (Gys1) was identified and conjugated to a cell penetrating peptide (GS-PPMO) to facilitate PMO delivery to muscle. GS-PPMO systemic administration to Pompe mice led to a dose-dependent decrease in glycogen synthase transcripts in the quadriceps, and the diaphragm but not the liver. An mRNA response in the heart was seen only at the higher dose tested. Associated with these decreases in transcript levels were correspondingly lower tissue levels of muscle specific glycogen synthase and activity. Importantly, these reductions resulted in significant decreases in the aberrant accumulation of lysosomal glycogen in the quadriceps, diaphragm, and heart of Pompe mice. Treatment was without any overt toxicity, supporting the notion that substrate reduction by GS-PPMO-mediated inhibition of muscle specific glycogen synthase represents a viable therapeutic strategy for Pompe disease after further development.

8.
Nucleic Acid Ther ; 23(2): 109-17, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23308382

RESUMEN

Expansions of CUG trinucleotide sequences in RNA transcripts provide the basis for toxic RNA gain-of-function that leads to detrimental changes in RNA metabolism. A CTG repeat element normally resides in the 3' untranslated region of the dystrophia myotonica-protein kinase (DMPK) gene, but when expanded it is the genetic lesion of myotonic dystrophy type 1 (DM1), a hereditary neuromuscular disease. The pathogenic DMPK transcript containing the CUG expansion is retained in ribonuclear foci as part of a complex with RNA-binding proteins such as muscleblind-like 1 (MBNL1), resulting in aberrant splicing of numerous RNA transcripts and consequent physiological abnormalities including myotonia. Herein, we demonstrate molecular and physiological amelioration of the toxic effects of mutant RNA in the HSA(LR) mouse model of DM1 by systemic administration of peptide-linked morpholino (PPMO) antisense oligonucleotides bearing a CAG repeat sequence. Intravenous administration of PPMO conjugates to HSA(LR) mice led to redistribution of Mbnl1 protein in myonuclei and corrections in abnormal RNA splicing. Additionally, myotonia was completely eliminated in PPMO-treated HSA(LR) mice. These studies provide proof of concept that neutralization of RNA toxicity by systemic delivery of antisense oligonucleotides that target the CUG repeat is an effective therapeutic approach for treating the skeletal muscle aspects of DM1 pathology.


Asunto(s)
Morfolinos/administración & dosificación , Distrofia Miotónica/genética , Péptidos/administración & dosificación , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3'/genética , Animales , Humanos , Ratones , Morfolinos/química , Mutación , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica , Oligonucleótidos Antisentido/administración & dosificación , Péptidos/química , Proteínas Serina-Treonina Quinasas/genética , ARN/genética , ARN/toxicidad , Empalme del ARN/genética , Expansión de Repetición de Trinucleótido/genética , Repeticiones de Trinucleótidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA