Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cancers (Basel) ; 15(15)2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37568579

RESUMEN

Parathyroid hormone-related peptide (PTHrP) is the primary cause of malignancy-associated hypercalcemia (MAH). We previously showed that PTHrP ablation, in the MMTV-PyMT murine model of breast cancer (BC) progression, can dramatically prolong tumor latency, slow tumor growth, and prevent metastatic spread. However, the signaling mechanisms using lineage tracing have not yet been carefully analyzed. Here, we generated Pthrpflox/flox; Cre+ mT/mG mice (KO) and Pthrpwt/wt; Cre+ mT/mG tumor mice (WT) to examine the signaling pathways under the control of PTHrP from the early to late stages of tumorigenesis. GFP+ mammary epithelial cells were further enriched for subsequent RNA sequencing (RNAseq) analyses. We observed significant upregulation of cell cycle signaling and fatty acid metabolism in PTHrP WT tumors, which are linked to tumor initiation and progression. Next, we observed that the expression levels of a novel lncRNA, GM50337, along with stearoyl-Coenzyme A desaturase 1 (Scd1) are significantly upregulated in PTHrP WT but not in KO tumors. We further validated a potential human orthologue lncRNA, OLMALINC, together with SCD1 that can be regulated via PTHrP in human BC cell lines. In conclusion, these novel findings could be used to develop targeted strategies for the treatment of BC and its metastatic complications.

2.
J Clin Invest ; 132(24)2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36326820

RESUMEN

The Hippo pathway nuclear effector Yes-associated protein (YAP) potentiates the progression of polycystic kidney disease (PKD) arising from ciliopathies. The mechanisms underlying the increase in YAP expression and transcriptional activity in PKD remain obscure. We observed that in kidneys from mice with juvenile cystic kidney (jck) ciliopathy, the aberrant hyperactivity of mechanistic target of rapamycin complex 1 (mTORC1), driven by ERK1/2 and PI3K/AKT cascades, induced ER proteotoxic stress. To reduce this stress by reprogramming translation, the protein kinase R-like ER kinase-eukaryotic initiation factor 2α (PERK/eIF2α) arm of the integrated stress response (ISR) was activated. PERK-mediated phosphorylation of eIF2α drove the selective translation of activating transcription factor 4 (ATF4), potentiating YAP expression. In parallel, YAP underwent K63-linked polyubiquitination by SCF S-phase kinase-associated protein 2 (SKP2) E3 ubiquitin ligase, a Hippo-independent, nonproteolytic ubiquitination that enhances YAP nuclear trafficking and transcriptional activity in cancer cells. Defective ISR cellular adaptation to ER stress in eIF2α phosphorylation-deficient jck mice further augmented YAP-mediated transcriptional activity and renal cyst growth. Conversely, pharmacological tuning down of ER stress/ISR activity and SKP2 expression in jck mice by administration of tauroursodeoxycholic acid (TUDCA) or tolvaptan impeded these processes. Restoring ER homeostasis and/or interfering with the SKP2-YAP interaction represent potential therapeutic avenues for stemming the progression of renal cystogenesis.


Asunto(s)
Proteínas Quinasas Asociadas a Fase-S , Ubiquitina-Proteína Ligasas , Ratones , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Factor de Transcripción Activador 4/metabolismo , Fosforilación , Riñón/metabolismo
3.
J Clin Endocrinol Metab ; 107(10): 2777-2783, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-35896139

RESUMEN

CONTEXT: Autosomal recessive hypophosphatemic rickets (ARHR) are rare, heritable renal phosphate-wasting disorders that arise from overexpression of the bone-derived phosphaturic hormone fibroblast growth factor 23 (FGF23) leading to impaired bone mineralization (rickets and osteomalacia). Inactivating mutations of Dentin matrix protein 1 (DMP1) give rise to ARHR type 1 (ARHR1). Short stature, prominent bowing of the legs, fractures/pseudofractures, and severe enthesopathy are prominent in this patient population. Traditionally, treatment consists of oral phosphate replacement and the addition of calcitriol but this approach is limited by modest efficacy and potential renal and gastrointestinal side effects. OBJECTIVE: The advent of burosumab (Crysvita), a fully humanized monoclonal antibody to FGF23 for the treatment of X-linked hypophosphatemia and tumor-induced osteomalacia, offers a unique opportunity to evaluate its safety and efficacy in patients with ARHR1. RESULTS: Monthly administration of burosumab to 2 brothers afflicted with the disorder resulted in normalization of serum phosphate, healing of pseudofracture, diminished fatigue, less bone pain, and reduced incapacity arising from the extensive enthesopathy and soft tissue fibrosis/calcification that characterizes this disorder. No adverse effects were reported following burosumab administration. CONCLUSION: The present report highlights the beneficial biochemical and clinical outcomes associated with the use of burosumab in patients with ARHR1.


Asunto(s)
Enfermedades Óseas Metabólicas , Entesopatía , Raquitismo Hipofosfatémico Familiar , Osteomalacia , Raquitismo Hipofosfatémico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Calcitriol/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/genética , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Hormonas/uso terapéutico , Humanos , Masculino , Osteomalacia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatémico/tratamiento farmacológico , Raquitismo Hipofosfatémico/genética
4.
Dev Neurobiol ; 81(1): 47-62, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33275829

RESUMEN

Parathyroid hormone-related peptide (PTHrP) acts under physiological conditions to regulate normal development of several tissues and organs. The role of PTHrP in spinal cord development has not been characterized. Pthrp knock in (Pthrp KI) mice were genetically modified to produce PTHrP in which there is a deficiency of the nuclear localization sequence (NLS) and C-terminus. Using this genetically modified mouse model, we have characterized its effect on spinal cord development early postnatally. The spinal cords from Pthrp KI mice displayed a significant reduction in its length, weight, and cross-sectional area compared to wild-type controls. Histologically, there was a decreased development of neurons and glial cells that caused decreased cell proliferation and increased apoptosis. The neural stem cells (NSCs) cultures also revealed decreased cell proliferation and differentiation and increased apoptosis. The proposed mechanism of delayed spinal cord development in Pthrp KI mice may be due to alteration in associated pathways in regulation of cell-division cycles and apoptosis. There was significant downregulation of Bmi-1 and upregulation of cyclin-dependent kinase inhibitors p27, p21, and p16 in Pthrp KI animals. We conclude that NLS and C-terminus peptide segments of PTHrP play an important role in inhibiting cell apoptosis and stimulation of cellular proliferation necessary for normal spinal cord development.


Asunto(s)
Apoptosis , Proteína Relacionada con la Hormona Paratiroidea , Animales , Núcleo Celular , Proliferación Celular , Ratones , Médula Espinal/fisiología
5.
Adv Exp Med Biol ; 1164: 161-178, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31576548

RESUMEN

PTHrP was first discovered as the most common mediator of malignancy-associated hypercalcemia. Subsequently, the discovery of its ubiquitous expression in normal tissues unraveled its role as a physiological autocrine/paracrine regulator. The significance of PTHrP in cancer is not confined to malignancy-associated hypercalcemia, and sufficient evidence now also supports its role in skeletal metastasis through its modulation of bone turnover. Furthermore, our own studies have recently shown the critical role of PTHrP in breast cancer initiation, growth, and metastasis. More recently, we have provided new evidence that overexpression of PTHrP is associated with higher incidence of brain metastasis and decreased overall survival in triple-negative breast cancer patients. Further mechanistic studies in human and mouse model are necessary to fully understand the role of PTHrP in tumor progression and metastasis.


Asunto(s)
Neoplasias Óseas , Hipercalcemia , Proteína Relacionada con la Hormona Paratiroidea , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/genética , Neoplasias Óseas/fisiopatología , Neoplasias de la Mama , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Hipercalcemia/etiología , Hipercalcemia/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo
6.
Sci Rep ; 7(1): 11208, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28894263

RESUMEN

Using a forward genetics approach to map loci in a mouse skin cancer model, we previously identified a genetic locus, Skin tumour modifier of MSM 1 (Stmm1) on chromosome 7, conferring strong tumour resistance. Sub-congenic mapping localized Parathyroid hormone (Pth) in Stmm1b. Here, we report that serum intact-PTH (iPTH) and a genetic polymorphism in Pth are important for skin tumour resistance. We identified higher iPTH levels in sera from cancer-resistant MSM/Ms mice compared with susceptible FVB/NJ mice. Therefore, we performed skin carcinogenesis experiments with MSM-BAC transgenic mice (Pth MSM-Tg) and Pth knockout heterozygous mice (Pth +/-). As a result, the higher amounts of iPTH in sera conferred stronger resistance to skin tumours. Furthermore, we found that the coding SNP (rs51104087, Val28Met) localizes in the mouse Pro-PTH encoding region, which is linked to processing efficacy and increased PTH secretion. Finally, we report that PTH increases intracellular calcium in keratinocytes and promotes their terminal differentiation. Taken together, our data suggest that Pth is one of the genes responsible for Stmm1, and serum iPTH could serve as a prevention marker of skin cancer and a target for new therapies.


Asunto(s)
Hormonas y Agentes Reguladores de Calcio/genética , Hormonas y Agentes Reguladores de Calcio/metabolismo , Predisposición Genética a la Enfermedad , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/genética , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , Polimorfismo de Nucleótido Simple
7.
Endocrinology ; 158(2): 252-263, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27929669

RESUMEN

Loss of fibroblast growth factor-23 (FGF23) causes hyperphosphatemia, extraskeletal calcifications, and early mortality; excess FGF23 causes hypophosphatemia with rickets or osteomalacia. However, FGF23 may not be important during fetal development. FGF23 deficiency (Fgf23 null) and FGF23 excess (Phex null) did not alter fetal phosphorus or skeletal parameters. In this study, we further tested our hypothesis that FGF23 is not essential for fetal phosphorus regulation but becomes important after birth. Although coreceptor Klotho null adults have extremely high FGF23 concentrations, intact FGF23 was normal in Klotho null fetuses, as were fetal phosphorus and skeletal parameters and placental and renal expression of FGF23 target genes. Pth/Fgf23 double mutants had the same elevation in serum phosphorus as Pth null fetuses, as compared with normal serum phosphorus in Fgf23 nulls. We examined the postnatal time courses of Fgf23 null, Klotho null, and Phex null mice. Fgf23 nulls and Klotho nulls were normal at birth, but developed hyperphosphatemia, increased renal expression of NaPi2a and NaPi2c, and reduced renal phosphorus excretion between 5 and 7 days after birth. Parathyroid hormone remained normal. In contrast, excess FGF23 exerted effects in Phex null males within 12 hours after birth, with the development of hypophosphatemia, reduced renal expression of NaPi2a and NaPi2c, and increased renal phosphorus excretion. In conclusion, although FGF23 is present in the fetal circulation at levels that may equal adult values, and there is robust expression of FGF23 target genes in placenta and fetal kidneys, FGF23 itself is not an important regulator of fetal phosphorous metabolism.


Asunto(s)
Feto/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Fósforo/sangre , Animales , Animales Recién Nacidos , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Glucuronidasa/genética , Glucuronidasa/metabolismo , Riñón/metabolismo , Proteínas Klotho , Masculino , Ratones Endogámicos C57BL , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Hormona Paratiroidea/sangre , Fenotipo , Embarazo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo
8.
J Clin Invest ; 126(2): 667-80, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26784541

RESUMEN

CYP24A1 (hereafter referred to as CYP24) enzymatic activity is pivotal in the inactivation of vitamin D metabolites. Basal renal and extrarenal CYP24 is usually low but is highly induced by its substrate 1,25-dihydroxyvitamin D. Unbalanced high and/or long-lasting CYP24 expression has been proposed to underlie diseases like chronic kidney disease, cancers, and psoriasis that otherwise should favorably respond to supplemental vitamin D. Using genetically modified mice, we have shown that renal phosphate wasting hypophosphatemic states arising from high levels of fibroblast growth factor 23 (FGF23) are also associated with increased renal Cyp24 expression, suggesting that elevated CYP24 activity is pivotal to the pathophysiology of these disorders. We therefore crossed 2 mouse strains, each with distinct etiology for high levels of circulating FGF23, onto a Cyp24-null background. Specifically, we evaluated Cyp24 deficiency in Hyp mice, the murine homolog of X-linked dominant hypophosphatemic rickets, and transgenic mice that overexpress a mutant FGF23 (FGF23R176Q) that is associated with the autosomal dominant form of hypophosphatemic rickets. Loss of Cyp24 in these murine models of human disease resulted in near-complete recovery of rachitic/osteomalacic bony abnormalities in the absence of any improvement in the serum biochemical profile. Moreover, treatment of Hyp and FGF23R1760-transgenic mice with the CYP24 inhibitor CTA102 also ameliorated their rachitic bones. Our results link CYP24 activity to the pathophysiology of FGF23-dependent renal phosphate wasting states and implicate pharmacologic CYP24 inhibition as a therapeutic adjunct for their treatment.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Factores de Crecimiento de Fibroblastos/metabolismo , Fosfatos/orina , Insuficiencia Renal Crónica , Vitamina D3 24-Hidroxilasa/antagonistas & inhibidores , Síndrome Debilitante , Animales , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Ratones Noqueados , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/orina , Vitamina D3 24-Hidroxilasa/genética , Vitamina D3 24-Hidroxilasa/metabolismo , Síndrome Debilitante/tratamiento farmacológico , Síndrome Debilitante/genética , Síndrome Debilitante/patología , Síndrome Debilitante/orina
9.
Endocrinology ; 156(8): 2774-80, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26052897

RESUMEN

The blood calcium concentration during fetal life is tightly regulated within a narrow range by highly interactive homeostatic mechanisms that include transport of calcium across the placenta and fluxes in and out of bone; the mechanisms of this regulation are poorly understood. Our findings that endochondral bone-specific PTH/PTHrP receptor (PPR) knockout (KO) mice showed significant reduction of fetal blood calcium concentration compared with that of control littermates at embryonic day 18.5 led us to focus on bone as a possibly major determinant of fetal calcium homeostasis. We found that the fetal calcium concentration of Runx2 KO mice was significantly higher than that of control littermates, suggesting that calcium flux into bone had a considerable influence on the circulating calcium concentration. Moreover, Runx2:PTH double mutant fetuses showed calcium levels similar to those of Runx2 KO mice, suggesting that part of the fetal hypocalcemia in PTH KO mice was caused by the increment of the mineralized bone mass allowed by the formation of osteoblasts. Finally, Rank:PTH double mutant mice had a blood calcium concentration even lower than that of the either Rank KO or PTH KO mice alone at embryonic day 18.5. These observations in our genetic models suggest that PTH/PTHrP receptor signaling in bones has a significant role of the regulation of fetal blood calcium concentration and that both placental transport and osteoclast activation contribute to PTH's hypercalcemic action. They also show that PTH-independent deposition of calcium in bone is the major controller of fetal blood calcium level.


Asunto(s)
Huesos/metabolismo , Calcio/sangre , Sangre Fetal/metabolismo , Homeostasis , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Hormona Paratiroidea/fisiología , Animales , Embrión de Mamíferos , Femenino , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Embarazo , Transducción de Señal/fisiología
10.
Am J Physiol Renal Physiol ; 305(3): F333-42, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23720345

RESUMEN

Parathyroid hormone-related protein (PTHrP) belongs to vasoactive factors that regulate blood pressure and renal hemodynamics both by reducing vascular tone and raising renin release. PTHrP is expressed in systemic and renal vasculature. Here, we wanted to assess the contribution of vascular smooth muscle cell endogenous PTHrP to the regulation of cardiovascular and renal functions. We generated a mouse strain (SMA-CreERT2/PTHrPL2/L2 or premutant PTHrPSM-/-), which allows temporally controlled, smooth muscle-targeted PTHrP knockdown in adult mice. Tamoxifen treatment induced efficient recombination of PTHrP-floxed alleles and decreased PTHrP expression in vascular and visceral smooth muscle cells of PTHrPSM-/- mice. Blood pressure remained unchanged in PTHrPSM-/- mice, but plasma renin concentration and creatinine clearance were reduced. Renal hemodynamics were further analyzed during clearance measurements in anesthetized mice. Conditional knockdown of PTHrP decreased renal plasma flow and glomerular filtration rate with concomitant reduction in filtration fraction. Similar measurements were repeated during acute saline volume expansion. Saline volume expansion induced a rise in renal plasma flow and reduced filtration fraction; both were blunted in PTHrPSM-/- mice leading to impaired diuresis. These findings show that endogenous vascular smooth muscle PTHrP controls renal hemodynamics under basal conditions, and it is an essential factor in renal vasodilation elicited by saline volume expansion.


Asunto(s)
Presión Sanguínea/genética , Presión Sanguínea/fisiología , Miocitos del Músculo Liso/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Circulación Renal/genética , Circulación Renal/fisiología , Animales , Antineoplásicos Hormonales/farmacología , Volumen Sanguíneo/fisiología , Cartilla de ADN , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inmunohistoquímica , Pruebas de Función Renal , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Musculares/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Renina/metabolismo , Tamoxifeno/farmacología
11.
Am J Physiol Endocrinol Metab ; 303(12): E1489-501, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23092913

RESUMEN

The human parathyroid hormone type 2 receptor (PTH2R) is activated by PTH and by tuberoinfundibular peptide of 39 residues (TIP39), the latter likely acting as its natural ligand. Although the receptor is expressed at highest levels in the nervous system, we have observed that both PTH2R and TIP39 are expressed in the newborn mouse growth plate, with the receptor localizing in the resting zone and the ligand TIP39 localizing exclusively in prehypertrophic and hypertrophic chondrocytes. To address the role of PTH2R in postnatal skeletal growth and development, Col2a1-hPTH2R (PTH2R-Tg) transgenic mice were generated. The mice were viable and of nearly normal size at birth. Expression of the transgene in the growth plate was limited to chondrocytes. We found that chondrocyte proliferation was decreased, as determined by in vivo BrdU labeling of proliferating chondrocytes and CDK4 and p21 expression in the growth plate of Col2a1-hPTH2R transgenic mice. Similarly, the differentiation and maturation of chondrocytes was delayed, as characterized by decreased Sox9 expression and weaker immunostaining for the chondrocyte differentiation markers collagen type II and type X and proteoglycans. As well, there was altered expression of Gdf5, Wdr5, and ß-catenin, factors implicated in chondrocyte maturation, proliferation, and differentiation.These effects impacted on the process of endochondral ossification, resulting in delayed formation of the secondary ossification center, and diminished trabecular bone volume. The findings substantiate a role for PTH2R signaling in postnatal growth plate development and subsequent bone mass acquisition.


Asunto(s)
Enfermedades del Desarrollo Óseo/metabolismo , Huesos/metabolismo , Condrocitos/metabolismo , Receptor de Hormona Paratiroídea Tipo 2/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Enfermedades del Desarrollo Óseo/patología , Huesos/patología , Diferenciación Celular , Proliferación Celular , Condrocitos/patología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Placa de Crecimiento/metabolismo , Placa de Crecimiento/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Oncogénica p21(ras)/metabolismo , Otosclerosis/metabolismo , Otosclerosis/patología , Receptor de Hormona Paratiroídea Tipo 2/biosíntesis , Receptor de Hormona Paratiroídea Tipo 2/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Factor de Transcripción SOX9/metabolismo , Vía de Señalización Wnt
12.
Am J Physiol Endocrinol Metab ; 302(7): E841-51, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22275754

RESUMEN

Although the calcium-sensing receptor (CaSR) and parathyroid hormone (PTH) may each exert skeletal effects, it is uncertain how CaSR and PTH interact at the level of bone in primary hyperparathyroidism (PHPT). Therefore, we simulated PHPT with 2 wk of continuous PTH infusion in adult mice with deletion of the PTH gene (Pth(-/-) mice) and with deletion of both PTH and CaSR genes (Pth(-/-)-Casr (-/-) mice) and compared skeletal phenotypes. PTH infusion in Pth(-/-) mice increased cortical bone turnover, augmented cortical porosity, and reduced cortical bone volume, femoral bone mineral density (BMD), and bone mineral content (BMC); these effects were markedly attenuated in PTH-infused Pth(-/-)-Casr(-/-) mice. In the absence of CaSR, the PTH-stimulated expression of receptor activator of nuclear factor-κB ligand and tartrate-resistant acid phosphatase and PTH-stimulated osteoclastogenesis was also reduced. In trabecular bone, PTH-induced increases in bone turnover, trabecular bone volume, and trabecular number were lower in Pth(-/-)-Casr(-/-) mice than in Pth(-/-) mice. PTH-stimulated genetic markers of osteoblast activity were also lower. These results are consistent with a role for CaSR in modulating both PTH-induced bone resorption and PTH-induced bone formation in discrete skeletal compartments.


Asunto(s)
Huesos/metabolismo , Hormona Paratiroidea/farmacología , Receptores Sensibles al Calcio/metabolismo , Absorciometría de Fotón , Fosfatasa Ácida/metabolismo , Animales , Densidad Ósea/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/patología , Células Cultivadas , Femenino , Fémur/diagnóstico por imagen , Regulación de la Expresión Génica , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Porosidad , ARN/biosíntesis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Sensibles al Calcio/genética , Fosfatasa Ácida Tartratorresistente , Tomografía Computarizada por Rayos X
13.
J Clin Invest ; 121(12): 4655-69, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22056386

RESUMEN

Parathyroid hormone-related protein (PTHrP) is a secreted factor expressed in almost all normal fetal and adult tissues. It is involved in a wide range of developmental and physiological processes, including serum calcium regulation. PTHrP is also associated with the progression of skeletal metastases, and its dysregulated expression in advanced cancers causes malignancy-associated hypercalcemia. Although PTHrP is frequently expressed by breast tumors and other solid cancers, its effects on tumor progression are unclear. Here, we demonstrate in mice pleiotropic involvement of PTHrP in key steps of breast cancer - it influences the initiation and progression of primary tumors and metastases. Pthrp ablation in the mammary epithelium of the PyMT-MMTV breast cancer mouse model caused a delay in primary tumor initiation, inhibited tumor progression, and reduced metastasis to distal sites. Mechanistically, it reduced expression of molecular markers of cell proliferation (Ki67) and angiogenesis (factor VIII), antiapoptotic factor Bcl-2, cell-cycle progression regulator cyclin D1, and survival factor AKT1. PTHrP also influenced expression of the adhesion factor CXCR4, and coexpression of PTHrP and CXCR4 was crucial for metastatic spread. Importantly, PTHrP-specific neutralizing antibodies slowed the progression and metastasis of human breast cancer xenografts. Our data identify what we believe to be new functions for PTHrP in several key steps of breast cancer and suggest that PTHrP may constitute a novel target for therapeutic intervention.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia/genética , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Proteínas Angiogénicas/biosíntesis , Proteínas Angiogénicas/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/fisiología , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/inmunología , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Receptores CXCR4/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Adv Exp Med Biol ; 720: 145-60, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21901625

RESUMEN

Parathyroid hormone-related protein (PTHrP) is widely expressed in fetal and adult tissues and is a key regulator for cellular calcium transport and smooth muscle cell contractility, as well as a crucial control factor in cell proliferation, development and differentiation. PTHrP stimulates or inhibits apoptosis in an autocrine/paracrine and intracrine fashion, and is particularly important for hair follicle and bone development, mammary epithelial development and tooth eruption. PTHrP's dysregulated expression has traditionally been associated with oncogenic pathologies as the major causative agent of malignancy-associated hypercalcemia, but recent evidence revealed a driving role in skeletal metastasis progression. Here, we demonstrate that PTHrP is also closely involved in breast cancer initiation, growth and metastasis through mechanisms separate from its bone turnover action, and we suggest that PTHrP as a facilitator of oncogenes would be a novel target for therapeutic purposes.


Asunto(s)
Neoplasias de la Mama/etiología , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Animales , Neoplasias Óseas/secundario , Mama/crecimiento & desarrollo , Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Proteína Relacionada con la Hormona Paratiroidea/química , Proteína Relacionada con la Hormona Paratiroidea/genética
15.
J Bone Miner Res ; 26(6): 1242-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21308774

RESUMEN

Mice lose 20% to 25% of trabecular bone mineral content (BMC) during lactation and restore it after weaning through unknown mechanisms. We found that tibial Pthrp mRNA expression was upregulated fivefold by 7 days after weaning versus end of lactation in wild-type (WT) mice. To determine whether parathyroid hormone-related protein (PTHrP) stimulates bone formation after weaning, we studied a conditional knockout in which PTHrP is deleted from preosteoblasts and osteoblasts by collagen I promoter-driven Cre (Cre(ColI) ). These mice are osteopenic as adults but have normal serum calcium, calcitriol, and parathyroid hormone (PTH). Pairs of Pthrp(flox/flox) ;Cre(ColI) (null) and WT;Cre(ColI) (WT) females were mated and studied through pregnancy, lactation, and 3 weeks of postweaning recovery. By end of lactation, both genotypes lost lumbar spine BMC: WT declined by 20.6% ± 3.3%, and null decreased by 22.5% ± 3.5% (p < .0001 versus baseline; p = NS between genotypes). During postweaning recovery, both restored BMC to baseline: WT to -3.6% ± 3.7% and null to 0.3% ± 3.7% (p = NS versus baseline or between genotypes). Similar loss and full recovery of BMC were seen at the whole body and hind limb. Histomorphometry confirmed that nulls had lower bone mass at baseline and that this was equal to the value achieved after weaning. Osteocalcin, propeptide of type 1 collagen (P1NP), and deoxypyridinoline increased equally during recovery in WT and null mice; PTH decreased and calcitriol increased equally; serum calcium was unchanged. Urine calcium increased during recovery but remained no different between genotypes. Although osteoblast-derived PTHrP is required to maintain adult bone mass and Pthrp mRNA upregulates in bone after weaning, it is not required for recovery of bone mass after lactation. The factors that stimulate postweaning bone formation remain unknown.


Asunto(s)
Huesos/fisiología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Destete , Animales , Fenómenos Biomecánicos/fisiología , Densidad Ósea/fisiología , Remodelación Ósea/fisiología , Calcitriol/sangre , Calcio/orina , Femenino , Regulación del Desarrollo de la Expresión Génica , Lactancia/sangre , Ratones , Osteoblastos/metabolismo , Hormona Paratiroidea/sangre , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Fósforo/orina , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducción/fisiología , Tibia/fisiología , Regulación hacia Arriba/genética
16.
Am J Physiol Endocrinol Metab ; 296(1): E79-88, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18984852

RESUMEN

Transgenic mice overexpressing fibroblast growth factor (FGF23) (R176Q) (F(Tg)) exhibit biochemical {hypophosphatemia, phosphaturia, abnormal 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] metabolism} and skeletal (rickets and osteomalacia) abnormalities attributable to FGF23 action. In vitro studies now implicate the aging-related factor Klotho in the signaling mechanism of FGF23. In this study, we used a mouse genetic approach to validate in vivo the pivotal role of Klotho in the metabolic and skeletal derangements associated with FGF23 (R176Q) overexpression. To this end, we crossed mice heterozygous for the hypomorphic Klotho allele (Kl(+/-)) to F(Tg) mice and obtained F(Tg) transgenic mice homozygous for the Kl-hypomorphic allele (F(Tg)/Kl(-/-)). Mice were killed on postnatal day 50, and serum and tissues were procured for analysis and comparison with F(Tg), wild-type, and Kl(-/-) controls. From 4 wk onward, F(Tg)/Kl(-/-) mice were clearly distinguishable from F(Tg) mice and exhibited a striking phenotypic resemblance to the Kl(-/-) controls. Serum analysis for calcium, phosphorus, parathyroid hormone, 1,25(OH)(2)D(3), and alkaline phosphatase activity confirmed the biochemical similarity between the F(Tg)/Kl(-/-) and Kl(-/-) mice and their distinctness from the F(Tg) controls. The characteristic skeletal changes associated with FGF23 (R176Q) overexpression were also dramatically reversed by the absence of Klotho. Hence the wide, unmineralized growth plates and the osteomalacic abnormalities apparent in trabecular and cortical bone were completely reversed in the F(Tg)/Kl(-/-) mice. Nevertheless, independent actions of Klotho on bone were suggested as manifested by alterations in mineralized bone, and in cortical bone volume which were observed in both the Kl(-/-) and F(Tr)/Kl(-/-) mutants. In summary, our findings substantiate in vivo the essential role of Klotho in the mechanism of action of FGF23 in view of the fact that Klotho ablation converts the biochemical and skeletal manifestations resulting from FGF23 overexpression to a phenotype consistent with Klotho deficiency.


Asunto(s)
Remodelación Ósea/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/deficiencia , Osteomalacia/metabolismo , Fosfatasa Ácida/sangre , Fosfatasa Alcalina/sangre , Animales , Northern Blotting , Calcitriol/sangre , Calcio/sangre , Cruzamientos Genéticos , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/genética , Glucuronidasa/genética , Glucuronidasa/metabolismo , Inmunohistoquímica , Isoenzimas/sangre , Proteínas Klotho , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteomalacia/sangre , Osteomalacia/genética , Osteomalacia/patología , Hormona Paratiroidea/sangre , Fenotipo , Fósforo/sangre , Fosfatasa Ácida Tartratorresistente
17.
Proc Natl Acad Sci U S A ; 105(51): 20309-14, 2008 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-19091948

RESUMEN

Parathyroid hormone (PTH) plays a central role in the regulation of serum calcium and phosphorus homeostasis, while parathyroid hormone-related protein (PTHrP) has important developmental roles. Both peptides signal through the same G protein-coupled receptor, the PTH/PTHrP or PTH type 1 receptor (PTH1R). PTHrP, normally a secreted protein, also contains a nuclear localization signal (NLS) that in vitro imparts functionality to the protein at the level of the nucleus. We investigated this functionality in vivo by introducing a premature termination codon in Pthrp in ES cells and generating mice that express PTHrP (1-84), a truncated form of the protein that is missing the NLS and the C-terminal region of the protein but can still signal through its cell surface receptor. Mice homozygous for the knock-in mutation (Pthrp KI) displayed retarded growth, early senescence, and malnutrition leading postnatally to their rapid demise. Decreased cellular proliferative capacity and increased apoptosis in multiple tissues including bone and bone marrow cells were associated with altered expression and subcellular distribution of the senescence-associated tumor suppressor proteins p16(INK4a) and p21 and the oncogenes Cyclin D, pRb, and Bmi-1. These findings provide in vivo experimental proof that substantiates the biologic relevance of the NLS and C-terminal portion of PTHrP, a polypeptide ligand that signals mainly via a cell surface G protein-coupled receptor.


Asunto(s)
Trastornos del Crecimiento/etiología , Señales de Localización Nuclear/deficiencia , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Animales , Apoptosis , Células de la Médula Ósea/patología , Huesos/patología , Proliferación Celular , Técnicas de Sustitución del Gen , Trastornos del Crecimiento/genética , Desnutrición/etiología , Desnutrición/genética , Ratones , Proteínas Mutantes , Oncogenes , Tasa de Supervivencia , Proteínas Supresoras de Tumor
18.
Eur J Endocrinol ; 158(5): 755-64, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18426836

RESUMEN

OBJECTIVE: Mice with osteoblast-specific deletion of parathyroid hormone-related protein (PTHrP) exhibit impaired recruitment and increased apoptosis of osteogenic cells resulting in decreased bone formation and premature osteoporosis. The PTHrP levels within the bone microenvironment are therefore critical in influencing bone mass acquisition. Whether this is applicable in humans has not been established. Here, we studied the association of a variable number of tandem repeats (VNTR) polymorphism in PTHrP with peak bone mass. METHODS: Enrolled in the study were 234 young Finnish males, with median age of 19.6 years (range 18.3-20.6 years). Lifestyle factors, serum bone markers, osteodensitometric measurements (lumbar spine and hip) and calcaneal quantitative ultrasound readings were obtained. The PTHrP VNTR length was determined by the PCR amplification of genomic DNA extracted from peripheral blood and correlated to bone parameters by the multiple regression models. RESULTS: The presence of at least one 252 bp allele was associated with increased lumbar spine bone mineral density (BMD; P<0.0034), broadband ultrasound attenuation (BUA; P<0.0012) and speed-of-sound (SOS; P<0.0023) measurements. The correlation with increased lumbar spine BMD (P=0.0008), BUA (P=0.005) and SOS (P=0.001) was further strengthened by the pairing of the 252 bp allele with a 460 bp allele in comparison with those without any 252 bp allele. Electrophoretic mobility shift assays were used to illustrate the potential transcriptional functionality of the VNTR sequence. CONCLUSION: The results indicate that the PTHrP VNTR sequence likely modulates local PTHrP expression within the skeletal microenvironment and could serve as a diagnostic predictor of peak bone mass acquisition.


Asunto(s)
Densidad Ósea/genética , Osteoporosis/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Secuencias Repetidas en Tándem , Adolescente , Adulto , Alelos , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Finlandia , Variación Genética , Humanos , Vértebras Lumbares/diagnóstico por imagen , Masculino , Datos de Secuencia Molecular , Osteoblastos/citología , Osteoblastos/fisiología , Osteoporosis/epidemiología , Polimorfismo Genético , Valor Predictivo de las Pruebas , Factores de Riesgo , Ultrasonografía
19.
Endocrinology ; 148(6): 2613-21, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332067

RESUMEN

Osteoporosis is a leading public health problem. Although a major cause in women is thought to be a decline in estrogen, it has recently been proposed that FSH or follitropin is required for osteoporotic bone loss. We examined the FSH receptor null mouse (FORKO mouse) to determine whether altered ovarian function could induce bone loss independent of FSH action. By 3 months of age, FORKO mice developed age-dependent declines in bone mineral density and trabecular bone volume of the lumbar spine and femur, which could be partly reversed by ovarian transplantation. Bilateral ovariectomy reduced elevated circulating testosterone levels in FORKO mice and decreased bone mass to levels indistinguishable from those in ovariectomized wild-type controls. Androgen receptor blockade and especially aromatase inhibition each produced bone volume reductions in the FORKO mouse. The results indicate that ovarian secretory products, notably estrogen, and peripheral conversion of ovarian androgen to estrogen can alter bone homeostasis independent of any bone resorptive action of FSH.


Asunto(s)
Huesos/fisiología , Hormona Folículo Estimulante/fisiología , Homeostasis , Ovario/fisiología , Andrógenos/farmacología , Animales , Densidad Ósea/genética , Huesos/efectos de los fármacos , Huesos/metabolismo , Femenino , Homeostasis/genética , Masculino , Ratones , Ratones Noqueados , Osteoporosis/patología , Ovariectomía , Ovario/fisiopatología , Ovario/trasplante , Receptores de HFE/genética
20.
J Clin Invest ; 115(9): 2402-11, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16138191

RESUMEN

Mice heterozygous for targeted disruption of Pthrp exhibit, by 3 months of age, diminished bone volume and skeletal microarchitectural changes indicative of advanced osteoporosis. Impaired bone formation arising from decreased BM precursor cell recruitment and increased apoptotic death of osteoblastic cells was identified as the underlying mechanism for low bone mass. The osteoporotic phenotype was recapitulated in mice with osteoblast-specific targeted disruption of Pthrp, generated using Cre-LoxP technology, and defective bone formation was reaffirmed as the underlying etiology. Daily administration of the 1-34 amino-terminal fragment of parathyroid hormone (PTH 1-34) to Pthrp+/- mice resulted in profound improvement in all parameters of skeletal microarchitecture, surpassing the improvement observed in treated WT littermates. These findings establish a pivotal role for osteoblast-derived PTH-related protein (PTHrP) as a potent endogenous bone anabolic factor that potentiates bone formation by altering osteoblast recruitment and survival and whose level of expression in the bone microenvironment influences the therapeutic efficacy of exogenous PTH 1-34.


Asunto(s)
Huesos , Osteoblastos/metabolismo , Osteogénesis/fisiología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Apoptosis/fisiología , Densidad Ósea , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Huesos/anatomía & histología , Huesos/citología , Huesos/metabolismo , Marcación de Gen , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/citología , Osteoporosis/patología , Osteoporosis/fisiopatología , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/genética , Hormona Paratiroidea/uso terapéutico , Proteína Relacionada con la Hormona Paratiroidea/genética , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA