Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(16): 7973-7981, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30926667

RESUMEN

Whole-body metabolic homeostasis is tightly controlled by hormone-like factors with systemic or paracrine effects that are derived from nonendocrine organs, including adipose tissue (adipokines) and liver (hepatokines). Fibroblast growth factor 21 (FGF21) is a hormone-like protein, which is emerging as a major regulator of whole-body metabolism and has therapeutic potential for treating metabolic syndrome. However, the mechanisms that control FGF21 levels are not fully understood. Herein, we demonstrate that FGF21 production in the liver is regulated via a posttranscriptional network consisting of the CCR4-NOT deadenylase complex and RNA-binding protein tristetraprolin (TTP). In response to nutrient uptake, CCR4-NOT cooperates with TTP to degrade AU-rich mRNAs that encode pivotal metabolic regulators, including FGF21. Disruption of CCR4-NOT activity in the liver, by deletion of the catalytic subunit CNOT6L, increases serum FGF21 levels, which ameliorates diet-induced metabolic disorders and enhances energy expenditure without disrupting bone homeostasis. Taken together, our study describes a hepatic CCR4-NOT/FGF21 axis as a hitherto unrecognized systemic regulator of metabolism and suggests that hepatic CCR4-NOT may serve as a target for devising therapeutic strategies in metabolic syndrome and related morbidities.


Asunto(s)
Exorribonucleasas , Factores de Crecimiento de Fibroblastos , Hepatocitos , Homeostasis , Ribonucleasas , Animales , Células Cultivadas , Dieta Alta en Grasa , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Hepatocitos/fisiología , Homeostasis/genética , Homeostasis/fisiología , Humanos , Hígado/química , Hígado/metabolismo , Hígado/patología , Síndrome Metabólico/metabolismo , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleasas/genética , Ribonucleasas/metabolismo
2.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055125

RESUMEN

PTH induces phosphorylation of the transcriptional coregulator NACA on serine 99 through Gαs and PKA. This leads to nuclear translocation of NACA and expression of the target gene Lrp6, encoding a coreceptor of the PTH receptor (PTH1R) necessary for full anabolic response to intermittent PTH (iPTH) treatment. We hypothesized that maintaining enough functional PTH1R/LRP6 coreceptor complexes at the plasma membrane through NACA-dependent Lrp6 transcription is important to ensure maximal response to iPTH. To test this model, we generated compound heterozygous mice in which one allele each of Naca and Lrp6 is inactivated in osteoblasts and osteocytes, using a knock-in strain with a Naca99 Ser-to-Ala mutation and an Lrp6 floxed strain (test genotype: Naca99S/A; Lrp6+/fl;OCN-Cre). Four-month-old females were injected with vehicle or 100 µg/kg PTH(1-34) once daily, 5 days a week for 4 weeks. Control mice showed significant increases in vertebral trabecular bone mass and biomechanical properties that were abolished in compound heterozygotes. Lrp6 expression was reduced in compound heterozygotes vs. controls. The iPTH treatment increased Alpl and Col1a1 mRNA levels in the control but not in the test group. These results confirm that NACA and LRP6 form part of a common genetic pathway that is necessary for the full anabolic effect of iPTH.


Asunto(s)
Anabolizantes/administración & dosificación , Células Madre Embrionarias/citología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Chaperonas Moleculares/genética , Hormona Paratiroidea/administración & dosificación , Anabolizantes/farmacología , Animales , Línea Celular , Membrana Celular/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Ratones , Chaperonas Moleculares/metabolismo , Mutagénesis Sitio-Dirigida , Osteoblastos/metabolismo , Osteocitos/metabolismo , Hormona Paratiroidea/farmacología , Fosforilación , Transducción de Señal/efectos de los fármacos , Microtomografía por Rayos X
3.
J Cell Physiol ; 236(2): 1195-1213, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32686190

RESUMEN

The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is a pleiotropic enzyme involved in DNA repair, cell cycle control, and transcription regulation. A potential role for DNA-PKcs in the regulation of osteoblastogenesis remains to be established. We show that pharmacological inhibition of DNA-PKcs kinase activity or gene silencing of Prkdc (encoding DNA-PKcs) in murine osteoblastic MC3T3-E1 cells and human adipose-derived mesenchymal stromal cells markedly enhanced osteogenesis and the expression of osteoblast differentiation marker genes. Inhibition of DNA-PKcs inhibited cell cycle progression and increased osteogenesis by significantly enhancing the bone morphogenetic protein 2 response in osteoblasts and other mesenchymal cell types. Importantly, in vivo pharmacological inhibition of the kinase enhanced bone biomechanical properties. Bones from osteoblast-specific conditional Prkdc-knockout mice exhibited a similar phenotype of increased stiffness. In conclusion, DNA-PKcs negatively regulates osteoblast differentiation, and therefore DNA-PKcs inhibitors may have therapeutic potential for bone regeneration and metabolic bone diseases.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Osteogénesis/genética , Animales , Dominio Catalítico/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Connect Tissue Res ; 62(2): 176-182, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-31462087

RESUMEN

Purpose/Aim of study: We previously cloned Tlcd3b2 (Tram-Lag1-CLN8 domain 3B2, formerly Fam57b2) from bone fracture repair callus tissue of Cyp24a1 knockout mice and showed that it synthesizes lactosylceramide (LacCer) under allosteric control of the vitamin D metabolite, 24,25-dihydroxyvitamin D3 [24,25(OH)2D3]. Tlcd3b2 was mainly detected in chondrocytes and the 24,25(OH)2D3-TLCD3B2-LacCer signaling cascade was shown to be important for optimal bone fracture repair, suggesting a role for TLCD3B2 in chondrocyte differentiation or maturation. We report the subcellular localization of TLCD3B2 and its effect on chondrocyte differentiation. Materials and Methods: Immunofluorescence detection of epitope-tagged mutants was used to assess localization. ATDC5 chondrogenic cells were transfected with Tlcd3b2 expression vectors to examine effects on chondrocyte differentiation. Results and Conclusions: TLCD3B2 localized to the endoplasmic reticulum, with both the N- and C-termini facing the cytosolic compartment. Chondrogenic ATDC5 cells stably overexpressing Tlcd3b2 showed elevated type 2 (Col2a1) and type 10 (Col10a1) collagen gene expression and increased proteoglycan synthesis, and the effect on Col2a1 was enhanced by treatment with 24,25(OH)2D3. LacCer treatment of ATDC5 cells potentiated Col10a1 expression. Our results show that TLCD3B2 is an ER protein and implicate its expression and enzymatic product in chondrocyte maturation.


Asunto(s)
Condrocitos , Animales , Diferenciación Celular , Células Cultivadas , Condrogénesis , Fracturas Óseas , Lactosilceramidos , Proteínas de la Membrana , Ratones
5.
Int J Mol Sci ; 22(14)2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34299363

RESUMEN

The ubiquitin-proteasome system regulates biological processes in normal and diseased states. Recent investigations have focused on ubiquitin-dependent modifications and their impacts on cellular function, commitment, and differentiation. Ubiquitination is reversed by deubiquitinases, including ubiquitin-specific peptidases (USPs), whose roles have been widely investigated. In this review, we explore recent findings highlighting the regulatory functions of USPs in osteoblasts and providing insight into the molecular mechanisms governing their actions during bone formation. We also give a brief overview of our work on USP53, a target of PTH in osteoblasts and a regulator of mesenchymal cell lineage fate decisions. Emerging evidence addresses questions pertaining to the complex layers of regulation exerted by USPs on osteoblast signaling. We provide a short overview of our and others' understanding of how USPs modulate osteoblastogenesis. However, further studies using knockout mouse models are needed to fully understand the mechanisms underpinning USPs actions.


Asunto(s)
Osteoblastos/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Linaje de la Célula/fisiología , Humanos , Activación de Linfocitos/fisiología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Transducción de Señal/fisiología , Ubiquitinación/fisiología
6.
J Biol Chem ; 294(20): 8184-8196, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30948508

RESUMEN

The transcriptional cofactor nascent polypeptide-associated complex and co-regulator α (NACA) regulates osteoblast maturation and activity. NACA functions, at least in part, by binding to Jun proto-oncogene, AP-1 transcription factor subunit (cJUN) and potentiating the transactivation of AP-1 targets such as osteocalcin (Bglap) and matrix metallopeptidase 9 (Mmp9). NACA activity is modulated by phosphorylation carried out by several kinases, but a phosphatase regulating NACA's activity remains to be identified. Here, we used affinity purification with MS in HEK293T cells to isolate NACA complexes and identified protein phosphatase 1 catalytic subunit α (PP1A) as a NACA-associated Ser/Thr phosphatase. NACA interacted with multiple components of the PP1A holoenzyme complex: the PPP1CA catalytic subunit and the regulatory subunits PPP1R9B, PPP1R12A and PPP1R18. MS analysis revealed that NACA co-expression with PPP1CA causes dephosphorylation of NACA at Thr-89, Ser-151, and Thr-174. NACA Ser/Thr-to-alanine variants displayed increased nuclear localization, and NACA dephosphorylation was associated with specific recruitment of novel NACA interactants, such as basic transcription factor 3 (BTF3) and its homolog BTF3L4. NACA and PP1A cooperatively potentiated cJUN transcriptional activity of the AP-1-responsive MMP9-luciferase reporter, which was abolished when Thr-89, Ser-151, or Thr-174 were substituted with phosphomimetic aspartate residues. We confirmed the NACA-PP1A interaction in MC3T3-E1 osteoblastic cells and observed that NACA phosphorylation status at PP1A-sensitive sites is important for the regulation of AP-1 pathway genes and for osteogenic differentiation and matrix mineralization. These results suggest that PP1A dephosphorylates NACA at specific residues, impacting cJUN transcriptional activity and osteoblast differentiation and function.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Osteoblastos/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular/genética , Animales , Núcleo Celular/genética , Células HEK293 , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Chaperonas Moleculares/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Fosforilación/genética , Proteína Fosfatasa 1/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-jun/genética , Elementos de Respuesta , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
J Immunol ; 199(12): 3952-3958, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29109124

RESUMEN

The vitamin D receptor participates in the control of IgE class-switch recombination in B cells. The physiologic vitamin D receptor agonist, 1,25(OH)2D3 (calcitriol), is synthesized by the essential enzyme 25-hydroxyvitamin D3-1α-hydroxylase (CYP27B1), which can be expressed by activated immune cells. The role of endogenous calcitriol synthesis for the regulation of IgE has not been proven. In this study, we investigated IgE-responses in Cyp27b1-knockout (KO) mice following sensitization to OVA or intestinal infection with Heligmosomoides polygyrus Specific Igs and plasmablasts were determined by ELISA and ELISpot, Cyp27b1 expression was measured by quantitative PCR. The data show elevated specific IgE and IgG1 concentrations in the blood of OVA-sensitized Cyp27b1-KO mice compared with wild-type littermates (+898 and +219%). Accordingly, more OVA-specific IgG1-secreting cells are present in spleen and fewer in the bone marrow of Cyp27b1-KO mice. Ag-specific mechanisms are suggested as the leucopoiesis is in general unchanged and activated murine B and T lymphocytes express Cyp27b1 Accordingly, elevated specific IgE concentrations in the blood of sensitized T cell-specific Cyp27b1-KO mice support a lymphocyte-driven mechanism. In an independent IgE-inducing model, i.e., intestinal infection with H. polygyrus, we validated the increase of total and specific IgE concentrations of Cyp27b1-KO compared with wild-type mice, but not those of IgG1 or IgA. We conclude that endogenous calcitriol has an impact on the regulation of IgE in vivo. Our data provide genetic evidence supporting previous preclinical and clinical findings and suggest that vitamin D deficiency not only promotes bone diseases but also type I sensitization.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/fisiología , Calcitriol/inmunología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/sangre , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/deficiencia , Animales , Linfocitos B/inmunología , Médula Ósea/inmunología , Calcitriol/biosíntesis , Calcitriol/deficiencia , Femenino , Helmintiasis Animal/inmunología , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Parasitosis Intestinales/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/inmunología , Especificidad de Órganos , Ovalbúmina/inmunología , Receptores de Calcitriol/fisiología , Bazo/inmunología , Linfocitos T/inmunología , Deficiencia de Vitamina D/inmunología
8.
J Am Soc Nephrol ; 28(2): 586-597, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27535551

RESUMEN

Circulating levels of fibroblast growth factor 23 (FGF23) increase during the early stages of kidney disease, but the underlying mechanism remains incompletely characterized. We investigated the role of vitamin D metabolites in regulating intact FGF23 production in genetically modified mice without and with adenine-induced uremia. Exogenous calcitriol (1,25-dihydroxyvitamin D) and high circulating levels of calcidiol (25-hydroxyvitamin D) each increased serum FGF23 levels in wild-type mice and in mice with global deficiency of the Cyp27b1 gene encoding 25-hydroxyvitamin D 1-α-hydroxylase, which produces 1,25-hydroxyvitamin D. Compared with wild-type mice, normal, or uremic mice lacking Cyp27b1 had lower levels of serum FGF23, despite having high concentrations of parathyroid hormone, but administration of exogenous 1,25-dihydroxyvitamin D increased FGF23 levels. Furthermore, raising serum calcium levels in Cyp27b1-depleted mice directly increased FGF23 levels and indirectly enhanced the action of ambient vitamin D metabolites via the vitamin D receptor. In chromatin immunoprecipitation assays, 25-hydroxyvitamin D promoted binding of the vitamin D receptor and retinoid X receptor to the promoters of osteoblastic target genes. Conditional osteoblastic deletion of Cyp27b1 caused lower serum FGF23 levels, despite normal circulating levels of vitamin D metabolites. In adenine-induced uremia, only a modest increase in serum FGF23 levels occurred in mice with osteoblastic deletion of Cyp27b1 (12-fold) compared with a large increase (58-fold) in wild-type mice. Therefore, in addition to the direct effect of high circulating concentrations of 25-hydroxyvitamin D, local osteoblastic conversion of 25-hydroxyvitamin D to 1,25-dihydroxyvitamin D appears to be an important positive regulator of FGF23 production, particularly in uremia.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Osteoblastos/metabolismo , Vitamina D/análogos & derivados , Animales , Factor-23 de Crecimiento de Fibroblastos , Enfermedades Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Vitamina D/biosíntesis , Vitamina D/fisiología
9.
Kidney Int ; 88(5): 1013-29, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26176830

RESUMEN

Vitamin D hydroxylated at carbon 25 (25(OH)D) is generally recognized as a precursor of active vitamin D. Despite its low affinity for the vitamin D receptor (VDR), both deficient and excessive 25(OH)D levels are associated with poor clinical outcomes. Here we studied direct effects of 25(OH)D3 on the kidney using 25(OH)D-1α-hydroxylase (CYP27B1) knockout mice. The effects of 25(OH)D3 on unilateral ureteral obstruction were analyzed as proximal tubular cells and macrophages are two major cell types that take up 25(OH)D and contribute to the pathogenesis of kidney injury. Excess 25(OH)D3 in obstructed mice worsened oxidative stress and tubulointerstitial fibrosis, whereas moderate levels of 25(OH)D3 had no effects. The exacerbating effects of excess 25(OH)D3 were abolished in CYP27B1/VDR double-knockout mice and in macrophage-depleted CYP27B1 knockout mice. Excess 25(OH)D3 upregulated both M1 marker (TNF-α) and M2 marker (TGF-ß1) levels of kidney-infiltrating macrophages. In vitro analyses verified that excess 25(OH)D3 directly upregulated TNF-α and TGF-ß1 in cultured macrophages but not in tubular cells. TNF-α and 25(OH)D3 cooperatively induced oxidative stress by upregulating iNOS in tubular cells. Aggravated tubulointerstitial fibrosis in mice with excess 25(OH)D3 indicated that macrophage-derived TGF-ß1 also had a key role in the pathogenesis of surplus 25(OH)D3. Thus, excess 25(OH)D3 worsens tubulointerstitial injury by modulating macrophage phenotype.


Asunto(s)
Calcifediol/farmacología , Túbulos Renales/patología , Macrófagos/metabolismo , Estrés Oxidativo/efectos de los fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Calcifediol/administración & dosificación , Calcifediol/metabolismo , Células Cultivadas , Femenino , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Receptores de Calcitriol/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Obstrucción Ureteral/complicaciones
10.
J Cell Biochem ; 115(5): 866-73, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24375853

RESUMEN

The transcriptional coregulator αNAC (Nascent polypeptide associated complex And Coregulator alpha) and the transcriptional repressor FIAT (Factor Inhibiting ATF4-mediated Transcription) interact but the biological relevance of this interaction remains unclear. The activity of αNAC is extensively modulated by post-translational modifications (PTMs). We identified a novel αNAC PTM through covalent attachment of the Small Ubiquitin-like MOdifier (SUMO1). Recombinant αNAC was a SUMO1 target in in vitro SUMOylation assays and we confirmed that αNAC is conjugated to SUMO1 in cultured osteoblasts and in calvarial tissue. The amino acid sequence of αNAC contains one copy of the composite "phospho-sumoyl switch" motif that couples sequential phosphorylation and SUMOylation. We found that αNAC is selectively SUMOylated at lysine residue 127 within the motif and that SUMOylation is enhanced when a phosphomimetic mutation is introduced at the nearby serine residue 132. SUMOylation did not alter the DNA-binding capacity of αNAC. The S132D, hyper-SUMOylated αNAC mutant specifically interacted with histone deacetylase-2 (HDAC2) and enhanced the inhibitory activity of FIAT on ATF4-mediated transcription from the Osteocalcin gene promoter. This effect required binding of SUMOylated αNAC to the target promoter. We propose that maximal transcriptional repression by FIAT requires its interaction with SUMOylated, HDAC2-interacting αNAC.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Sumoilación/genética , Transcripción Genética , Animales , Proteínas Co-Represoras/genética , Histona Desacetilasa 2/metabolismo , Ratones , Chaperonas Moleculares/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Fosforilación/genética , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional/genética
11.
Am J Pathol ; 182(4): 1099-106, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23375622

RESUMEN

Osteoarthritis (OA) is an age-related progressive degenerative joint disease. Peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor, is suggested as an attractive therapeutic target to counteract degradative mechanisms associated with OA. Studies suggest that activation of PPARγ by its agonists can reduce the synthesis of OA catabolic and inflammatory factors and the development of cartilage lesions in OA animal models. Because these agonists impart several PPARγ-independent effects, the specific in vivo function of PPARγ in cartilage homeostasis and OA remains largely unknown. Herein, we describe the in vivo role of PPARγ in OA using cartilage-specific PPARγ knockout (KO) mice generated using the Cre-lox system. Adult PPARγ KO mice exhibited a spontaneous OA phenotype associated with enhanced cartilage degradation, hypocellularity, synovial and cartilage fibrosis, synovial inflammation, mononuclear cell influx in the synovium, and increased expression of catabolic factors, including matrix metalloproteinase-13, accompanied by an increase in staining for matrix metalloproteinase-generated aggrecan and type II collagen neoepitopes (VDIPEN and C1-2C). We demonstrate that PPARγ-deficient articular cartilage exhibits elevated expression of the additional catabolic factors hypoxia-inducible factor-2α, syndecan-4, and a disintegrin and metalloproteinase with thrombospondin motifs 5 and of the inflammatory factors cyclooxygenase-2 and inducible nitric oxide synthase. In conclusion, PPARγ is a critical regulator of cartilage health, the lack of which leads to an accelerated spontaneous OA phenotype.


Asunto(s)
Envejecimiento/metabolismo , Cartílago/metabolismo , Cartílago/patología , Osteoartritis/metabolismo , Osteoartritis/patología , PPAR gamma/deficiencia , Animales , Biomarcadores/metabolismo , Fibrosis , Eliminación de Gen , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Especificidad de Órganos , PPAR gamma/metabolismo , Fenotipo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
12.
JBMR Plus ; 8(5): ziae012, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38577520

RESUMEN

Calcitriol circulates at low levels in normal human and rodent fetuses, in part due to increased 24-hydroxylation of calcitriol and 25-hydroxyvitamin D by 24-hydroxylase (CYP24A1). Inactivating mutations of CYP24A1 cause high postnatal levels of calcitriol and the human condition of infantile hypercalcemia type 1, but whether the fetus is disturbed by the loss of CYP24A1 is unknown. We hypothesized that loss of Cyp24a1 in fetal mice will cause high calcitriol, hypercalcemia, and increased placental calcium transport. The Cyp24a1+/- mice were mated to create pregnancies with wildtype, Cyp24a1+/-, and Cyp24a1 null fetuses. The null fetuses were hypercalcemic, modestly hypophosphatemic (compared to Cyp24a1+/- fetuses only), with 3.5-fold increased calcitriol, 4-fold increased fibroblast growth factor 23 (FGF23), and unchanged parathyroid hormone. The quantitative RT-PCR confirmed the absence of Cyp24a1 and 2-fold increases in S100g, sodium-calcium exchanger type 1, and calcium-sensing receptor in null placentas but not in fetal kidneys; these changes predicted an increase in placental calcium transport. However, placental 45Ca and 32P transport were unchanged in null fetuses. Fetal ash weight and mineral content, placental weight, crown-rump length, and skeletal morphology did not differ among the genotypes. Serum procollagen 1 intact N-terminal propeptide and bone expression of sclerostin and Blgap were reduced while calcitonin receptor was increased in nulls. In conclusion, loss of Cyp24a1 in fetal mice causes hypercalcemia, modest hypophosphatemia, and increased FGF23, but no alteration in skeletal development. Reduced incorporation of calcium into bone may contribute to the hypercalcemia without causing a detectable decrease in the skeletal mineral content. The results predict that human fetuses bearing homozygous or compound heterozygous inactivating mutations of CYP24A1 will also be hypercalcemic in utero but with normal skeletal development.

13.
J Bone Miner Res ; 39(5): 595-610, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38477809

RESUMEN

Ablation of Cyp27b1 eliminates calcitriol but does not disturb fetal mineral homeostasis or skeletal development. However, independent of fetal genotypes, maternal loss of Cyp27b1 altered fetal mineral and hormonal levels compared to offspring of WT dams. We hypothesized that these maternal influences would alter postnatal skeletal development. Cyp27b1 null and WT females were mated to bear only Cyp27b1+/- offspring. Forty-eight hours after birth, pups were cross-fostered to dams of the same or opposite genotype that bore them. Maternal and offspring samples were collected on days 21 (weaning) and 42. Offspring measurements included minerals and hormones, BMC by DXA, ash weight and mineral content, gene expression, 3-point bending tests, and microCT. Maternal lactational behavior was evaluated. Milk was analyzed for nutritional content. At day 21, offspring fostered by nulls, independent of birth dam, had ~20% lower weight, BMC, ash weight, and ash calcium than pups fostered by WT dams. Adjustment for body weight accounted for the lower BMC but not the lower ash weight and ash calcium. Hormones and serum/urine minerals did not differ across offspring groups. Offspring fostered by nulls had shorter femurs and lower cortical thickness, mean polar moment of inertia, cortical area, trabecular bone volume, and trabecular number. Dam lactational behaviors and milk nutritional content did not differ between groups. At day 42, body weight, ash weight, lengths, BMC, and tibial bone strength were no longer different between pups fostered by null vs WT dams. In summary, pups fostered by Cyp27b1 nulls, regardless of birth dam, have proportionately smaller skeletons at 21 d, impaired microstructure, but normal mineral homeostasis. The skeletal effects are largely recovered by day 42 (3 wk after weaning). In conclusion, maternal loss of calcitriol impairs early postnatal cortical bone growth and trabecular bone mass, but affected offspring catch up after weaning.


Asunto(s)
Desarrollo Óseo , Calcitriol , Animales , Femenino , Calcitriol/sangre , Calcitriol/metabolismo , Desarrollo Óseo/efectos de los fármacos , Ratones , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Densidad Ósea/efectos de los fármacos , Lactancia , Masculino , Embarazo , Ratones Noqueados , Peso Corporal/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/metabolismo
14.
Biochim Biophys Acta ; 1819(11-12): 1208-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23092676

RESUMEN

In the nucleus of differentiated osteoblasts, the DNA-binding αNAC protein acts as a transcriptional coactivator of the Osteocalcin gene. Chromatin immunoprecipitation-microarray assays (ChIP-chip) showed that αNAC binds the Osteocalcin promoter but also identified the Myogenin promoter as an αNAC target. Here, we confirm these array data using quantitative ChIP and further detected that αNAC binds to these promoters in myoblasts. Since these genes are differentially regulated during osteoblastogenesis or myogenesis, these results suggest cell- and promoter-context specific functions for αNAC. We hypothesized that αNAC dynamically recruits corepressors to inhibit Myogenin expression in cells committing to the osteoblastic lineage or to inhibit Osteocalcin transcription in differentiating myoblasts. Using co-immunoprecipitation assays, we detected complexes between αNAC and the corepressors HDAC1 and HDAC3, in myoblasts and osteoblasts. Sequential ChIP confirmed HDAC1 recruitment by αNAC at the Osteocalcin and Myogenin promoters. Interaction with the corepressors was detectable in pre-osteoblasts and in myoblasts but disappeared as the cells differentiate. Treatment with an HDAC inhibitor caused de-repression of Osteocalcin expression in myoblasts. Overexpression of αNAC in myoblasts inhibits expression of Myogenin and differentiation. However, overexpression of an N-terminus truncated αNAC mutant allowed myoblasts to express Myogenin and differentiate, and this mutant did not interact with HDAC1 or HDAC3. This study identified an additional DNA-binding target and novel protein-protein interactions for αNAC. We propose that αNAC plays a role in regulating gene transcription during mesenchymal cell differentiation by differentially recruiting corepressors at target promoters.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasas/metabolismo , Chaperonas Moleculares/metabolismo , Mioblastos/metabolismo , Miogenina/biosíntesis , Osteoblastos/metabolismo , Osteocalcina/biosíntesis , Regiones Promotoras Genéticas/fisiología , Animales , Diferenciación Celular/fisiología , Línea Celular , Histona Desacetilasa 1/genética , Histona Desacetilasas/genética , Ratones , Chaperonas Moleculares/genética , Mioblastos/citología , Miogenina/genética , Osteoblastos/citología , Osteocalcina/genética , Transcripción Genética/fisiología
15.
J Cell Biochem ; 114(8): 1863-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23463631

RESUMEN

FIAT (factor inhibiting ATF4-mediated transcription) represses Osteocalcin gene transcription and inhibits osteoblast activity by heterodimerizing with ATF4 to prevent it from binding DNA. It thus appears important to identify and characterize the molecular mechanisms that control Fiat gene expression in osteoblasts. In silico sequence analysis identified a canonical GC-box within a 1,400 bp region of the proximal Fiat gene promoter. Electrophoretic mobility shift assays (EMSA) with MC3T3-E1 osteoblastic cells nuclear extracts indicated that the transcription factors Sp1 and Sp3, but not Sp7/OSTERIX, bound this proximal GC-box. Chromatin immunoprecipitation confirmed interaction of the two transcription factors with the Fiat promoter GC-element in living osteoblasts. Transient transfection studies showed that Sp1 dose-dependently activated the expression of a Fiat-luciferase reporter construct while both the long or short isoforms of Sp3 dose-dependently inhibited transcription from the Fiat reporter construct. Transfection of an Sp7/OSTERIX expression vector did not affect expression of the Fiat-luciferase reporter. Co-transfection of increasing amounts of the Sp3 expression vector in the context of maximal Sp1-dependent Fiat-luciferase activation led to dose-dependent repression of the expression of the reporter. Using RNA knockdown, we measured a reduction in steady-state Fiat expression when Sp1 was inhibited, and a reciprocal increase upon Sp3 knockdown. In parallel, treatment of osteoblasts with WP631, which prevents Sp1/DNA interactions, strongly inhibited the expression of Fiat and reduced the occupancy of the Fiat promoter proximal GC-box by Sp1. Taken together, our results suggest an interplay between Sp1 and Sp3 as a mechanism involved in the control of Fiat gene expression in osteoblasts.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Elementos de Respuesta/fisiología , Factores de Transcripción Sp/metabolismo , Animales , Línea Celular , Proteínas Co-Represoras/genética , Ratones , Proteínas Nucleares/genética , Osteoblastos/citología , Factores de Transcripción Sp/genética
16.
Arthritis Rheum ; 64(5): 1551-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22131019

RESUMEN

OBJECTIVE: Long bones develop through the strictly regulated process of endochondral ossification within the growth plate, resulting in the replacement of cartilage by bone. Defects in this process can result in skeletal abnormalities and a predisposition to degenerative joint diseases such as osteoarthritis (OA). Studies suggest that activation of the transcription factor peroxisome proliferator-activated receptor γ (PPARγ) is an important therapeutic target in OA. To devise PPARγ-related therapies in OA, it is critical to identify the role of this transcription factor in cartilage biology. Therefore, this study sought to determine the in vivo role of PPARγ in endochondral ossification and cartilage development, using cartilage-specific PPARγ-knockout (KO) mice. METHODS: Cartilage-specific PPARγ-KO mice were generated using the Cre/loxP system. Histomorphometric and immunohistochemical analyses were performed to assess the patterns of ossification, proliferation, differentiation, and hypertrophy of chondrocytes, skeletal organization, bone density, and calcium deposition in the KO mice. RESULTS: PPARγ-KO mice exhibited reductions in body length, body weight, length of the long bones, skeletal growth, cellularity, bone density, calcium deposition, and trabecular bone thickness, abnormal organization of the growth plate, loss of columnar organization, shorter hypertrophic zones, and delayed primary and secondary ossification. Immunohistochemical analyses for Sox9, 5-bromo-2'-deoxyuridine, p57, type X collagen, and platelet endothelial cell adhesion molecule 1 revealed reductions in the differentiation, proliferation, and hypertrophy of chondrocytes and in vascularization of the growth plate in mutant mice. Isolated chondrocytes and cartilage explants from mutant mice showed aberrant expression of Sox9 and extracellular matrix markers, including aggrecan, type II collagen, and matrix metalloproteinase 13. In addition, chondrocytes from mutant mice exhibited enhanced phosphorylation of p38 and decreased expression of Indian hedgehog. CONCLUSION: The presence of PPARγ is required for normal endochondral ossification and cartilage development in vivo.


Asunto(s)
Huesos/patología , Cartílago/patología , Condrocitos/metabolismo , Condrogénesis/fisiología , Osteogénesis/fisiología , PPAR gamma/biosíntesis , Animales , Biomarcadores/metabolismo , Densidad Ósea , Desarrollo Óseo , Huesos/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Hipertrofia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/deficiencia , PPAR gamma/genética , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Bone Miner Res ; 38(4): 578-596, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36726200

RESUMEN

In the skeleton, osteoblasts and osteoclasts synchronize their activities to maintain bone homeostasis and integrity. Investigating the molecular mechanisms governing bone remodeling is critical and helps understand the underlying biology of bone disorders. Initially, we have identified the ubiquitin-specific peptidase gene (Usp53) as a target of the parathyroid hormone in osteoblasts and a regulator of mesenchymal stem cell differentiation. Mutations in USP53 have been linked to a constellation of developmental pathologies. However, the role of Usp53 in bone has never been visited. Here we show that Usp53 null mice have a low bone mass phenotype in vivo. Usp53 null mice exhibit a pronounced decrease in trabecular bone indices including trabecular bone volume (36%) and trabecular number (26%) along with an increase in trabecular separation (13%). Cortical bone parameters are also impacted, showing a reduction in cortical bone volume (12%) and cortical bone thickness (15%). As a result, the strength and mechanical bone properties of Usp53 null mice have been compromised. At the cellular level, the ablation of Usp53 perturbs bone remodeling, augments osteoblast-dependent osteoclastogenesis, and increases osteoclast numbers. Bone marrow adipose tissue volume increased significantly with age in Usp53-deficient mice. Usp53 null mice displayed increased serum receptor activator of NF-κB ligand (RANKL) levels, and Usp53-deficient osteoblasts and bone marrow adipocytes have increased expression of Rankl. Mechanistically, USP53 regulates Rankl expression by enhancing the interaction between VDR and SMAD3. This is the first report describing the function of Usp53 during skeletal development. Our results put Usp53 in display as a novel regulator of osteoblast-osteoclast coupling and open the door for investigating the involvement of USP53 in pathologies. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Médula Ósea , Osteoblastos , Ligando RANK , Proteasas Ubiquitina-Específicas , Animales , Ratones , Adipocitos/metabolismo , Huesos/metabolismo , Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Homeostasis , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Ligando RANK/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo
18.
Nutrients ; 14(15)2022 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-35956396

RESUMEN

Infantile hypercalcemia type 1 (HCINF1), previously known as idiopathic infantile hypercalcemia, is caused by mutations in the 25-hydroxyvitamin D 24-hydroxylase gene, CYP24A1. The R396W loss-of-function mutation in CYP24A1 is the second most frequent mutated allele observed in affected HCINF1 patients. We have introduced the site-specific R396W mutation within the murine Cyp24a1 gene in knock-in mice to generate a humanized model of HCINF1. On the C57Bl6 inbred background, homozygous mutant mice exhibited high perinatal lethality with 17% survival past weaning. This was corrected by crossbreeding to the CD1 outbred background. Mutant animals had hypercalcemia in the first week of life, developed nephrolithiasis, and had a very high 25(OH)D3 to 24,25(OH)2D3 ratio which is a diagnostic hallmark of the HCINF1 condition. Expression of the mutant Cyp24a1 allele was highly elevated while Cyp27b1 expression was abrogated. Impaired bone fracture healing was detected in CD1-R396w/w mutant animals. The augmented lethality of the C57Bl6-R396W strain suggests an influence of distinct genetic backgrounds. Our data point to the utility of unique knock-in mice to probe the physiological ramifications of CYP24A1 variants in isolation from other biological and environmental factors.


Asunto(s)
Hipercalcemia , Animales , Femenino , Homeostasis , Hipercalcemia/genética , Ratones , Minerales , Mutación , Embarazo , Vitamina D/metabolismo , Vitamina D3 24-Hidroxilasa/genética , Vitamina D3 24-Hidroxilasa/metabolismo , Vitaminas
19.
Nephron Exp Nephrol ; 119(3): e67-74, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21849802

RESUMEN

BACKGROUND/AIMS: Kidney disease patients experience declining calcitriol levels and develop secondary hyperparathyroidism (SHPT). Animal models of uremia based on 5/6 nephrectomy (NTX) do not consistently reproduce this calcitriol deficiency. We developed an animal model, the NTX Cyp27b1-null mouse, which completely lacks endogenous calcitriol, and examined the suitability of this model for evaluation of treatment with vitamin D analogs in uremia. METHODS: NTX was performed at 2 months of age. One week post-NTX, animals were treated for 4 weeks with vehicle; doxercalciferol at 30, 100 or 300 pg/g body weight (b.w.); or paricalcitol at 100, 300 or 1,000 pg/g b.w. by gavage 3 times per week. RESULTS: Serum blood urea nitrogen and creatinine were elevated. Vehicle-treated NTX null mice had hypocalcemia and SHPT. Doxercalciferol at 100 or 300 pg/g b.w. normalized serum calcium and parathyroid hormone (PTH) levels. Paricalcitol at 300 or 1,000 pg/g normalized serum calcium, but PTH levels remained elevated. Osteomalacia was corrected by 100 pg/g b.w. of doxercalciferol or 1,000 pg/g b.w. of paricalcitol. The highest dose of doxercalciferol, but not of paricalcitol, significantly reduced osteitis fibrosa. CONCLUSION: Our results reveal the differential efficacy of doxercalciferol and paricalcitol in this novel animal model incorporating both calcitriol deficiency and renal insufficiency.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/deficiencia , Ergocalciferoles/uso terapéutico , Uremia/tratamiento farmacológico , Uremia/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Administración Oral , Animales , Calcitriol/deficiencia , Calcitriol/metabolismo , Calcio/sangre , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ergocalciferoles/administración & dosificación , Ratones , Ratones Noqueados , Osteítis/tratamiento farmacológico , Osteítis/patología , Osteomalacia/tratamiento farmacológico , Osteomalacia/patología , Hormona Paratiroidea/sangre , Uremia/etiología
20.
Sci Rep ; 11(1): 8418, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33875709

RESUMEN

We have previously shown that parathyroid hormone (PTH) induces the phosphorylation of the DNA-binding protein Nascent polypeptide associated complex And Coregulator alpha (NACA), leading to nuclear translocation of NACA and activation of target genes. Using ChIP-Seq against NACA in parallel with RNA-sequencing, we report the identification of Ubiquitin Specific Peptidase 53 (Usp53) as a target gene of PTH-activated NACA in osteoblasts. A binding site for NACA within the ChIP fragment from the Usp53 promoter was confirmed by electrophoretic mobility shift assay. Activity of the Usp53 promoter (- 2325/+ 238 bp) was regulated by the JUN-CREB complex and this activation relied on activated PKA and the presence of NACA. Usp53 knockdown in ST2 stromal cells stimulated expression of the osteoblastic markers Bglap2 (Osteocalcin) and Alpl (Alkaline phosphatase) and inhibited expression of the adipogenic markers Pparg and Cebpa. A similar effect was measured when knocking down Naca. During osteoblastogenesis, the impact of Usp53 knockdown on PTH responses varied depending on the maturation stage of the cells. In vivo implantation of Usp53-knockdown bone marrow stromal cells in immunocompromised mice showed an increase in osteoblast number and a decrease in adipocyte counts. Our data suggest that Usp53 modulates the fate of mesenchymal cells by impacting lineage selection.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Adipogénesis , Fosfatasa Alcalina/metabolismo , Animales , Ratones , Osteocalcina/metabolismo , Hormona Paratiroidea/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA