Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Calcif Tissue Int ; 87(3): 263-72, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20563797

RESUMEN

Many patients with symptomatic bone metastases receive radiation therapy, even though radiation is known to have potential adverse effects on bone. We hypothesized that the concurrent use of a bisphosphonate drug (zoledronic acid, ZA) or a combination of ZA plus an anabolic agent (parathyroid hormone, PTH) would lead to improvements in the microarchitecture and mechanical properties of irradiated bone. Human breast cancer cells were injected into the distal femur of 56 female nude mice, which were then divided into four groups: no treatment (0 Gy), radiation administered 4 weeks postinjection (20 Gy), radiation plus ZA (12.5 microg/kg weekly from weeks 4 to 12) (20 Gy + ZA), and radiation followed by ZA (25 microg/kg weekly from weeks 4 to 8) and PTH(1-34) (100 microg microg/kg daily from weeks 8 to 12) (20 Gy + ZA + PTH). Left limbs served as normal control bones. Bone loss over the 12-week study was tracked with serial radiography and bone densitometry. At the end of the study, micro-computed tomography and mechanical testing were used to quantify bone microarchitecture and bone strength. Radiation alone failed to prevent tumor-induced decreases in bone mineral density (BMD), trabecular bone volume, and bone strength. Treatment with 20 Gy + ZA or 20 Gy + ZA + PTH as adjuncts to radiation was effective at preserving trabecular bone architecture and bone strength at normal levels. ZA reduced the risk of mechanical fragility following irradiation of a lytic bone lesion. Supplemental use of PTH did not result in further increases in bone strength but was associated with significant increases in BMD and bone mass, suggesting that it may be beneficial in enhancing bone architecture following radiation therapy.


Asunto(s)
Anabolizantes/administración & dosificación , Conservadores de la Densidad Ósea/administración & dosificación , Carcinoma/radioterapia , Difosfonatos/administración & dosificación , Neoplasias Femorales/radioterapia , Imidazoles/administración & dosificación , Osteólisis/prevención & control , Hormona Paratiroidea/administración & dosificación , Animales , Densidad Ósea/efectos de los fármacos , Huesos , Carcinoma/patología , Quimioterapia Combinada , Femenino , Neoplasias Femorales/patología , Ratones , Ratones Desnudos , Osteólisis/patología , Ácido Zoledrónico
2.
Endocrinology ; 145(6): 2815-23, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15016722

RESUMEN

G protein-coupled receptors (GPCRs) mediate the action of many hormones, cytokines, and sensory and chemical signals. It is generally thought that receptor desensitization and internalization require occupancy and activation of the GPCR. PTH and PTHrP receptor (PTH1R) belongs to GPCR class B and is the major regulator of extracellular calcium homeostasis. Using kidney distal convoluted tubule cells transfected with a human PTH1R/enhanced green fluorescent protein fusion protein, quantitative, real-time fluorescence microscopy was used to analyze receptor internalization. In these cells, which are the target of the calcium-sparing action of PTH, PTH(1-34) activated adenylyl cyclase (AC) and phospholipase C (PLC) and PTH1R endocytosis. PTH(1-31), however, stimulated AC and PLC but not PTH1R endocytosis. Conversely, PTH(7-34) rapidly stimulated PTH1R internalization without activating AC or PLC. PTH(2-34) and (3-34) caused PTH1R internalization intermediate between PTH(1-34) and (7-34). PTH1R sequestration occurred in a dynamin- and clathrin-dependent manner. Directly activating AC inhibited PTH1R internalization in response to PTH(7-34). PTH1R endocytosis was sensitive to protein kinase C inhibition. PTH(1-34), (7-34), and (1-31) evoked PTH1R phosphorylation. Removal of most of the C terminus of the PTH1R eliminated receptor phosphorylation and the cAMP/protein kinase C sensitivity of internalization. PTH(1-34) and (7-34) internalized the truncated PTH1R with identical kinetics, and the response was unaffected by forskolin. Thus, the PTH1R C terminus contains regulatory sequences that are involved in, but not required for, PTH1R internalization. The results demonstrate that receptor activation and internalization can be selectively dissociated.


Asunto(s)
Endocitosis , Receptor de Hormona Paratiroídea Tipo 1/fisiología , Animales , Caveolas/fisiología , Células Cultivadas , Clatrina/fisiología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Humanos , Membranas Intracelulares/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Ligandos , Ratones , Hormona Paratiroidea/química , Fragmentos de Péptidos/farmacología , Fosforilación , Fosfotransferasas/fisiología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptores de Hormona Paratiroidea/metabolismo , Receptores de Hormona Paratiroidea/fisiología , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal
3.
Crit Rev Eukaryot Gene Expr ; 12(1): 23-51, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12433064

RESUMEN

There is a need for anabolic drugs that can stimulate bone growth, improve bone microarchitecture, accelerate fracture healing and thus restore bone strength to oteoporotics. The anabolic agents currently leading the way to the clinic are the parathroid hormone (PTH) and some of its adenylyl cyclase-stimulating fragments. Here we discuss what is known about the genes and their products that are stimulated by PTHR1 receptor signals and in four ways cause a large accumulation of bone-building osteoblasts. We will also discuss the currently controversial anabolic activity of the cholesterol-lowering statins and outline a possible mechanism by which they might stimulate BMP-2 expession and bone growth. Finally, we will present the growing evidence for the body's "fat-o-stat" cytokine-leptin-indirectly restraining bone growth via a hypothalamic factor and at the same serving as a local autocrine/paracrine stimulator of osteoblast activity via IGF-I and an inhibitor of osteoclast generation by stimulating osteoblastic cells' antiosteoclast OPG (osteoprotegerin) expression and reducing their proosteoclast RANKL expression.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Desarrollo Óseo/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Leptina/fisiología , Hormona Paratiroidea/farmacología , Animales , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Complejos Multienzimáticos/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal
4.
Curr Med Chem ; 11(21): 2867-81, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15544480

RESUMEN

Osteogenic peptides are, or have potential to be, therapies for the treatment of osteoporosis, fracture repair, and repair of loosened bone implants. Human parathyroid hormone has been approved for the treatment of post-menopausal osteoporosis. Constrained analogs of PTH and the parathyroid-hormone related peptide (PTHrP) have aided the understanding of how PTH and PTHrP bind to their common receptor and some of these analogs have improved properties that make them possible candidates for clinical trial. Cyclization by lactam formation has shown that a core region of human PTH (hPTH) from residues 16-26 binds as an alpha-helix to the receptor and that the biological effects are remarkably sensitive to ring size. Appropriate cyclization in this region of the molecule not only has yielded analogs with improved receptor activation but also ones less susceptible to protease degradation and thus more active in vivo. Cyclization has been less successful in the N-terminus region, residues 1-12, of hPTH(1-34) with only a cyclization between residues 6 and 10 showing some promise. The growing understanding of how this region binds to the receptor will lead to other productive constraints. This review also covers the potential of a different class of molecule, the osteogenic growth peptide (OPG), as an anabolic bone agent. These molecules have much weaker anabolic effects than PTH and cyclization does not result in improved activity. However, the information gained from these studies may yield analogs with better pharmacological profiles.


Asunto(s)
Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/análogos & derivados , Hormona Paratiroidea/farmacología , Secuencia de Aminoácidos , Animales , Histonas , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/farmacología , Datos de Secuencia Molecular , Hormona Paratiroidea/química , Hormona Paratiroidea/uso terapéutico , Receptores de Hormona Paratiroidea/metabolismo
5.
J Med Chem ; 45(10): 2024-31, 2002 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-11985470

RESUMEN

In an effort to improve the activities and bioavailabilities of stromal cell-derived factor-1 (SDF-1, CXCL12) sdf-(1-67)-OH (1), we have prepared a linear peptide analogue [sdf-(1-31)-NH(2) (2)] and two lactam analogues [cyclo(Lys(20)-Glu(24))-sdf-(1-31)-NH(2) (3) and cyclo(Glu(24)-Lys(28))-sdf-(1-31)-NH(2) (4)], consisting of the N-terminal region (amino acids 1-14) joined by a four-glycine linker to the C-terminal region (amino acids 56-67) of 1. Analogues 2 and 4 had eight residues of alpha-helix, as estimated from its circular dichroism (CD) spectra, in contrast to 10 residues in analogue 3. Cyclization of analogue 2 at residues 20 and 24 to give analogue 3 resulted in only a slight change to the theta;(222)/theta;(209) ratio (0.81 to 0.86, where 1.09 is considered a perfect alpha-helix), although an increase in the alpha-helix length of analogue 3 was observed. In contrast, cyclization between residues 24 and 28 by lactamization to give analogue 4 only slightly affected the helical content but clearly resulted in a more classical alpha-helical structure (theta;(222)/theta;(209) = 0.98). Cyclization of the linear analogue 2 enhanced the SDF-1 receptor CXCR4 binding approximately 114-fold, where the IC(50) values derived from (125)I-SDF-1 competitive binding assays with CEM cells were found to be 39.5 +/- 5.9 nM, 28.9 +/- 6.3 microM, 225.8 +/- 11.8 nM, and 254.1 +/- 5.4 nM for analogues 1-4, respectively. Intracellular calcium mobilization ([Ca(2+)](i)) induced after interaction with CXCR4, as measured by EC(50), was significantly reduced in analogue 4 compared to 3, and approached the EC(50) of native SDF-1, indicating a correlation between the degree of alpha-helix and biological activity. Therefore, the biological activity of small peptide SDF-1 analogues is highly dependent on the conformation of its C-terminal region.


Asunto(s)
Quimiocinas CXC/química , Lactamas/síntesis química , Péptidos Cíclicos/síntesis química , Receptores CXCR4/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Línea Celular , Quimiocina CXCL12 , Dicroismo Circular , Humanos , Lactamas/química , Lactamas/farmacología , Datos de Secuencia Molecular , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Estructura Secundaria de Proteína , Ensayo de Unión Radioligante , Receptores CXCR4/agonistas
6.
Cancer Lett ; 200(2): 107-13, 2003 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-14568163

RESUMEN

The parathyroid hormone (PTH) and some of its fragments and analogs stimulate bone growth in various animal models and humans and one of them (hPTH-(1-34)) has been approved by the USFDA for treating osteoporosis. However, there are reports that PTH can stimulate the PI-3 kinase/mitogen-activated protein kinases-mediated proliferation of rat enterocytes and that primary hyperparathyroidism in humans is associated with an increased incidence of colon cancer. Here we have investigated the ability of two PTH fragments, hPTH-(1-34)NH(2) and [Leu(27)]cyclo(Glu(22)-Lys(26))hPTH-(1-31)NH(2) to initiate colon carcinogenesis or increase the initiatory activity of the widely used colon carcinogen azoxymethane (AOM). The initiation of colon carcinogenesis by AOM was indicated by the very early appearance of aberrant crypt foci. While both PTH peptides strongly stimulated femoral bone formation, they did not cause the appearance of ACFs or affect the number or the distribution along the colon of AOM-induced ACFs. Nor did AOM affect the PTHs' ability to stimulate bone formation. Thus, a relatively short PTH treatment that is long enough to strongly stimulate bone formation does not initiate colon carcinogenesis in rats.


Asunto(s)
Neoplasias del Colon/inducido químicamente , Hormona Paratiroidea/farmacología , Lesiones Precancerosas/inducido químicamente , Animales , Azoximetano , Carcinógenos , Osteogénesis , Fragmentos de Péptidos , Ratas , Ratas Sprague-Dawley
7.
Treat Endocrinol ; 1(3): 175-90, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-15799210

RESUMEN

The susceptibility to traumatic fracturing of osteopenic bones, and the spontaneous fracturing of osteoporotic bones by normal body movements caused by the microstructural deterioration and loss of bone, are currently treated with antiresorptive drugs, such as the bisphosphonates, calcitonin, estrogens, and selective estrogen receptor modulators. These antiresorptive agents target osteoclasts and, as their name indicates, reduce or stop bone resorption. They cannot directly stimulate bone formation, increase bone mass above normal values in ovariectomized rat models, or improve microstructure. However, there is a family of agents - the parathyroid hormone (PTH) and some of its fragments and their analogs - which directly stimulate bone growth and improve microstructure independently from impairing osteoclasts. These drugs are about to make their clinical debut in treating patients with osteoporosis and, probably not too far in the future, for accelerating fracture healing. They stimulate osteoblast accumulation and bone formation in three ways via signals from the type 1 PTH/PTH-related protein (PTHR1) receptors on proliferatively inactive preosteoblasts, osteoblasts, osteocytes and bone-lining cells. The receptor signals shut down the proliferative machinery in preosteoblasts and push their maturation to osteoblasts, cause the osteoblastic cells to make and secrete several factors that stimulate the extensive proliferation of osteoprogenitors without PTHRI receptors, stimulate the reversion of bone-lining cells to osteoblasts, and extend osteoblast lifespan and productivity by preventing them from suicidally initiating apoptosis. The first of the PTHs to reach the clinic will be teriparatide [recombinant human (h)PTH-(1-34)], which was recommended for approval in 2001 by the US Food and Drug Administration Endocrinology and Metabolic Drugs Advisory Committee for the treatment of postmenopausal osteoporosis. Teriparatide has been shown to considerably increase cancellous and cortical bone mass, improve bone microstructure, prevent fractures and thus provide benefits that cannot be provided by current antiresorptive drugs, when administered subcutaneously at a daily dose of 20 microg for no longer than 2 years to patients with osteoporosis.


Asunto(s)
Osteoporosis/tratamiento farmacológico , Hormona Paratiroidea/análogos & derivados , Hormona Paratiroidea/uso terapéutico , Animales , Densidad Ósea/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Ensayos Clínicos como Asunto , Terapia de Reemplazo de Estrógeno , Femenino , Humanos , Masculino , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporosis/patología , Fragmentos de Péptidos/uso terapéutico , Ratas , Receptores de Hormona Paratiroidea/metabolismo , Teriparatido/uso terapéutico
11.
Biochemistry ; 45(37): 11113-21, 2006 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16964972

RESUMEN

We have studied the effects of C-terminal group modifications (amide, methylamide, dimethylamide, aldehyde, and alcohol) on the conformation, adenylyl cyclase stimulation (AC), or binding of parathyroid hormone (hPTH) analogues, hPTH(1-28)NH(2) and hPTH(1-31)NH(2). hPTH(1-31)NH(2) has a C-terminal alpha-helix bounded by residues 17-29 [Chen, Z., et al. (2000) Biochemistry 39, 12766]. In both cases, relative to the natural analogue with a carboxyl C-terminus, the amide and methylamide had increased helix content whereas the dimethylamide forms had CD spectra more similar to the carboxyl one. Conformational effects were more pronounced with hPTH(1-28) than with hPTH(1-31), with increases in helix content of approximately 30% in contrast to 10%. Stabilization of the C-terminal helix of residues 1-28 seemed to correlate with an ability of the C-terminal function to H-bond appropriately. None of the analogues affected the AC stimulating activity significantly, but there was an up to 15-fold decrease in the level of apparent binding of the carboxyl hPTH(1-28) analogue compared to that of the methylamide and a 4-fold decrease in the level of binding of the aldehyde or dimethylamide. There was no significant change in binding activities for the 1-31 analogues. These observations are consistent with previous studies that imply the importance of a region of the hormone's C-terminal alpha-helix for tight binding to the receptor. They also show that modulation of helix stability does have an effect on the binding of the hormone, but only when the C-terminus is at the putative end of the helix. The similarity of AC stimulation even when binding changed 10-fold can be explained by assuming greater efficacy of the weaker binding PTH-receptor complexes in stimulating AC.


Asunto(s)
Hormona Paratiroidea/química , Fragmentos de Péptidos/química , Animales , Línea Celular , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Macaca mulatta , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Termodinámica
12.
J Biol Chem ; 281(43): 32485-95, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16923805

RESUMEN

The principal receptor-binding domain (Ser(17)-Val(31)) of parathyroid hormone (PTH) is predicted to form an amphiphilic alpha-helix and to interact primarily with the N-terminal extracellular domain (N domain) of the PTH receptor (PTHR). We explored these hypotheses by introducing a variety of substitutions in region 17-31 of PTH-(1-31) and assessing, via competition assays, their effects on binding to the wild-type PTHR and to PTHR-delNt, which lacks most of the N domain. Substitutions at Arg(20) reduced affinity for the intact PTHR by 200-fold or more, but altered affinity for PTHR-delNt by 4-fold or less. Similar effects were observed for Glu substitutions at Trp(23), Leu(24), and Leu(28), which together form the hydrophobic face of the predicted amphiphilic alpha-helix. Glu substitutions at Arg(25), Lys(26), and Lys(27) (which forms the hydrophilic face of the helix) caused 4-10-fold reductions in affinity for both receptors. Thus, the side chains of Arg(20), together with those composing the hydrophobic face of the ligand's putative amphiphilic alpha-helix, contribute strongly to PTHR-binding affinity by interacting specifically with the N domain of the receptor. The side chains projecting from the opposite helical face contribute weakly to binding affinity by different mechanisms, possibly involving interactions with the extracellular loop/transmembrane domain region of the receptor. The data help define the roles that side chains in the binding domain of PTH play in the PTH-PTHR interaction process and provide new clues for understanding the overall topology of the bimolecular complex.


Asunto(s)
Hormona Paratiroidea/química , Fragmentos de Péptidos/química , Receptores de Hormona Paratiroidea/química , Receptores de Hormona Paratiroidea/metabolismo , Alanina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Aminoácidos/química , Técnicas de Cultivo de Célula , Línea Celular , Dicroismo Circular , Ácido Glutámico/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
13.
J Biol Chem ; 280(25): 23771-7, 2005 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-15826940

RESUMEN

We have used backbone N-methylations of parathyroid hormone (PTH) to study the role of these NH groups in the C-terminal amphiphilic alpha-helix of PTH (1-31) in binding to and activating the PTH receptor (P1R). The circular dichroism (CD) spectra indicated the structure of the C-terminal alpha-helix was locally disrupted around the methylation site. The CD spectra differences were explained by assuming a helix disruption for four residues on each side of the site of methylation and taking into account the known dependence of CD on the length of an alpha-helix. Binding and adenylyl cyclase-stimulating data showed that outside of the alpha-helix, methylation of residues Asp30 and Val31 had little effect on structure or activities. Within the alpha-helix, disruption of the structure was associated with increased loss of activity, but for specific residues Val21, Leu24, Arg25, and Leu28 there was a dramatic loss of activities, thus suggesting a more direct role of these NH groups in correct P1R binding and activation. Activity analyses with P1R-delNT, a mutant with its long N-terminal region deleted, gave a different pattern of effects and implicated Ser17, Trp23, and Lys26 as important for its PTH activation. These two groups of residues are located on opposite sides of the helix. These results are compatible with the C-terminal helix binding to both the N-terminal segment and also to the looped-out extracellular region. These data thus provide direct evidence for important roles of the C-terminal domain of PTH in determining high affinity binding and activation of the P1R receptor.


Asunto(s)
Hormona Paratiroidea/metabolismo , Adenilil Ciclasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Dicroismo Circular , Humanos , Metilación , Datos de Secuencia Molecular , Hormona Paratiroidea/química , Porcinos
14.
Vitam Horm ; 65: 1-80, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12481542

RESUMEN

In the new millennium, humans will be traveling to Mars and eventually beyond with skeletons that respond to microgravity by self-destructing. Meanwhile in Earth's aging populations growing numbers of men and many more women are suffering from crippling bone loss. During the first decade after menopause all women suffer an accelerating loss of bone, which in some of them is severe enough to result in "spontaneous" crushing of vertebrae and fracturing of hips by ordinary body movements. This is osteoporosis, which all too often requires prolonged and expensive care, the physical and mental stress of which may even kill the patient. Osteoporosis in postmenopausal women is caused by the loss of estrogen. The slower development of osteoporosis in aging men is also due at least in part to a loss of the estrogen made in ever smaller amounts in bone cells from the declining level of circulating testosterone and is needed for bone maintenance as it is in women. The loss of estrogen increases the generation, longevity, and activity of bone-resorbing osteoclasts. The destructive osteoclast surge can be blocked by estrogens and selective estrogen receptor modulators (SERMs) as well as antiosteoclast agents such as bisphosphonates and calcitonin. But these agents stimulate only a limited amount of bone growth as the unaffected osteoblasts fill in the holes that were dug by the now suppressed osteoclasts. They do not stimulate osteoblasts to make bone--they are antiresorptives not bone anabolic agents. (However, certain estrogen analogs and bisphosphates may stimulate bone growth to some extent by lengthening osteoblast working lives.) To grow new bone and restore bone strength lost in space and on Earth we must know what controls bone growth and destruction. Here we discuss the newest bone controllers and how they might operate. These include leptin from adipocytes and osteoblasts and the statins that are widely used to reduce blood cholesterol and cardiovascular damage. But the main focus of this article is necessarily the currently most promising of the anabolic agents, the potent parathyroid hormone (PTH) and certain of its 31- to 38-aminoacid fragments, which are either in or about to be in clinical trial or in the case of Lilly's Forteo [hPTH-(1-34)] tentatively approved by the Food and Drug Administration for treating osteoporosis and mending fractures.


Asunto(s)
Huesos/lesiones , Fracturas Óseas/tratamiento farmacológico , Osteoporosis/tratamiento farmacológico , Animales , Desarrollo Óseo , Remodelación Ósea , Estrógenos , Femenino , Sustancias de Crecimiento , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Osteogénesis , Osteoporosis Posmenopáusica/prevención & control , Hormona Paratiroidea
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA