Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pancreatology ; 21(7): 1299-1304, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34446336

RESUMEN

BACKGROUND: The calcium sensing receptor (CASR) is a G protein-coupled receptor that is responsible for assessing extracellular Ca2+ levels and thus plays a crucial role in calcium homeostasis. Hypercalcemia is a metabolic risk factor for pancreatitis and rare CASR variants have been described in patients with chronic pancreatitis. At the carboxy-terminal tail of CASR, there is a cluster of three common polymorphisms, p.A986S (rs1801725), p.R990G (rs1042636) and p.Q1011E (rs1801726), which have been associated with chronic pancreatitis in various studies, but with conflicting results. METHODS: We examined 542 German and 339 French patients with chronic pancreatitis as well as 1025 German controls for the 3 common CASR polymorphism by melting curve analysis. For comparison, we used genotype data from 583 French controls from a previous study. In addition, we functionally analyzed the three variants by NFAT and SRE luciferase reporter systems as well as Western blotting and verified cell surface expression by ELISA. RESULTS: In both cohorts, neither the genotype nor the allele frequencies differed significantly between patients and controls. In both luciferase assays, p.R990G showed a significant leftward shift, indicating an increased responsiveness of the receptor. p.A986S showed a leftward shift in the SRE but not in the NFAT reporter assay, while the responsiveness of p.Q1011E did not differ from the wild-type. These functional studies therefore do not support the contributions of variant CASR to increasing the risk of pancreatitis. CONCLUSIONS: The three frequent CASR polymorphisms are unlikely to increase the risk for chronic pancreatitis.


Asunto(s)
Pancreatitis Crónica , Receptores Sensibles al Calcio , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación , Pancreatitis Crónica/genética , Polimorfismo Genético , Receptores Sensibles al Calcio/genética , Adulto Joven
2.
Semin Cell Dev Biol ; 49: 37-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26612442

RESUMEN

The calcium-sensing receptor (CaSR) expressed in the parathyroid gland and the kidney tubule acts as the calciostat and orchestrates blood calcium homeostasis by modulating production and release of parathyroid hormone (PTH) and active vitamin D that influence Ca(2+) fluxes across the bone, kidney and intestine. Here we consider the role of the CaSR as a responder to proinflammatory cytokines released as part of the innate immune response to tissue injury and inflammation with resetting of the calciostat on the one hand and as a promoter and mediator of the initial inflammatory response on the other. The importance of the CaSR in systemic calcium homeostasis is exemplified by the fact that inactivating and activating mutations in the gene result in hypercalcemia and hypocalcemia, respectively. Proinflammatory cytokines interleukin-1ß and interleukin-6 upregulate CaSR expression in parathyroid and kidney and do this through defined response elements in the CASR gene promoters. This results in decreased serum PTH and 1,25-dihydroxyvitamin D and calcium levels. This is likely to underlie the hypocalcemia that commonly occurs in critically ill patients, those with burn injury and sepsis, for example. The level of calcium in extracellular fluid bathing necrotic cells is often elevated and acts as a chemokine to attract monocytes/macrophages that express the CaSR to sites of tissue injury. Elevated levels of calcium acting via the CaSR can function as a danger signal that stimulates assembly of myeloid cell cytosolic multiprotein inflammasomes resulting in maturation of the proinflammatory cytokine IL-1ß by caspase-1. Thus the CaSR is both promoter of and responder to the inflammation.


Asunto(s)
Calcio/metabolismo , Citocinas/fisiología , Receptores Sensibles al Calcio/fisiología , Animales , Secuencia de Bases , Expresión Génica , Homeostasis , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo
3.
J Biol Chem ; 291(5): 2043-54, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26598524

RESUMEN

TGFß is a multifunctional cytokine that regulates cell proliferation, cell immortalization, and cell death, acting as a key homeostatic mediator in various cell types and tissues. Autophagy is a programmed mechanism that plays a pivotal role in controlling cell fate and, consequently, many physiological and pathological processes, including carcinogenesis. Although autophagy is often considered a pro-survival mechanism that renders cells viable in stressful conditions and thus might promote tumor growth, emerging evidence suggests that autophagy is also a tumor suppressor pathway. The relationship between TGFß signaling and autophagy is context-dependent and remains unclear. TGFß-mediated activation of autophagy has recently been suggested to contribute to the growth inhibitory effect of TGFß in hepatocarcinoma cells. In the present study, we define a novel process of TGFß-mediated autophagy in cancer cell lines of various origins. We found that autophagosome initiation and maturation by TGFß is dependent on the retinoblastoma tumor suppressor protein/E2 promoter binding factor (pRb/E2F1) pathway, which we have previously established as a critical signaling axis leading to various TGFß tumor suppressive effects. We further determined that TGFß induces pRb/E2F1-dependent transcriptional activation of several autophagy-related genes. Together, our findings reveal that TGFß induces autophagy through the pRb/E2F1 pathway and transcriptional activation of autophagy-related genes and further highlight the central relevance of the pRb/E2F1 pathway downstream of TGFß signaling in tumor suppression.


Asunto(s)
Autofagia , Carcinoma Hepatocelular/metabolismo , Factor de Transcripción E2F1/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína de Retinoblastoma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Células Hep G2 , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología
4.
J Biol Chem ; 290(7): 3910-24, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25538250

RESUMEN

Menin, the product of the multiple endocrine neoplasia type 1 (Men1) tumor suppressor gene, mediates the cell proliferation and differentiation actions of transforming growth factor-ß (TGF-ß) ligand family members. In vitro, menin modulates osteoblastogenesis and osteoblast differentiation promoted and sustained by bone morphogenetic protein-2 (BMP-2) and TGF-ß, respectively. To examine the in vivo function of menin in bone, we conditionally inactivated Men1 in mature osteoblasts by crossing osteocalcin (OC)-Cre mice with floxed Men1 (Men1(f/f)) mice to generate mice lacking menin in differentiating osteoblasts (OC-Cre;Men1(f/f) mice). These mice displayed significant reduction in bone mineral density, trabecular bone volume, and cortical bone thickness compared with control littermates. Osteoblast and osteoclast number as well as mineral apposition rate were significantly reduced, whereas osteocyte number was increased. Primary calvarial osteoblasts proliferated more quickly but had deficient mineral apposition and alkaline phosphatase activity. Although the mRNA expression of osteoblast marker and cyclin-dependent kinase inhibitor genes were all reduced, that of cyclin-dependent kinase, osteocyte marker, and pro-apoptotic genes were increased in isolated Men1 knock-out osteoblasts compared with controls. In contrast to the knock-out mice, transgenic mice overexpressing a human menin cDNA in osteoblasts driven by the 2.3-kb Col1a1 promoter, showed a gain of bone mass relative to control littermates. Osteoblast number and mineral apposition rate were significantly increased in the Col1a1-Menin-Tg mice. Therefore, osteoblast menin plays a key role in bone development, remodeling, and maintenance.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/fisiología , Diferenciación Celular , Osteoblastos/citología , Proteínas Proto-Oncogénicas/fisiología , Animales , Apoptosis , Western Blotting , Densidad Ósea , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Osteoblastos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
5.
BMC Cancer ; 15: 200, 2015 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-25885043

RESUMEN

BACKGROUND: Cutaneous melanoma is the most lethal skin cancer and its incidence in developed countries has dramatically increased over the past decades. Localized tumors are easily treated by surgery, but advanced melanomas lack efficient treatment and are associated with very poor outcomes. Thus, understanding the processes underlying melanoma development and progression is critical. The Transforming Growth Factor beta (TGFß) acts as a potent tumor suppressor in human melanoma, by inhibiting cell growth and preventing cellular migration and invasion. METHODS: In this study, we aimed at elucidating the molecular mechanisms underlying TGFß-mediated tumor suppression. Human cutaneous melanoma cell lines, derived from different patients, were used to assess for cell cycle analysis, apoptosis/caspase activity and cell migration. Techniques involved immunoblotting, immunohistochemistry, real time PCR and luciferase reporter assays. RESULTS: We found the leukemia inhibitory factor (LIF) to be strongly up-regulated by TGFß in melanoma cells, defining LIF as a novel TGFß downstream target gene in cutaneous melanoma. Interestingly, we also showed that TGFß-mediated LIF expression is required for TGFß-induced cell cycle arrest and caspase-mediated apoptosis, as well as for TGFß-mediated inhibition of cell migration. Moreover, we found that TGFß-mediated LIF expression leads to activation of transcription of the cell cycle inhibitor p21 in a STAT3-dependent manner, and further showed that p21 is required for TGFß/LIF-mediated cell cycle arrest and TGFß-induced gene activation of several pro-apoptotic genes. CONCLUSIONS: Together, our results define the LIF/p21 signaling cascade as a novel tumor suppressive-like pathway in melanoma, acting downstream of TGFß to regulate cell cycle arrest and cell death, further highlight new potential therapeutic strategies for the treatment of cutaneous melanoma.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor Inhibidor de Leucemia/biosíntesis , Melanoma/genética , Neoplasias Cutáneas/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Movimiento Celular/genética , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor Inhibidor de Leucemia/genética , Masculino , Melanoma/metabolismo , Melanoma/patología , Persona de Mediana Edad , Cultivo Primario de Células , Unión Proteica , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factor de Crecimiento Transformador beta1/metabolismo , Melanoma Cutáneo Maligno
6.
Breast Cancer Res ; 16(6): 476, 2014 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-25499443

RESUMEN

INTRODUCTION: This study helps to define the implications of breast cancer anti-estrogen resistance 3 (BCAR3) in breast cancer and extends the current understanding of its molecular mechanism of action. BCAR3 has been shown to promote cell proliferation, migration and attachment to extracellular matrix components. However, in a cohort of metastatic breast cancer patients who received tamoxifen treatment, high BCAR3 mRNA levels were associated with favorable progression-free survival outcome. These results suggest that, besides its established roles, BCAR3 may have additional mechanisms of action that regulate breast cancer aggressive phenotype. In this study, we investigated whether BCAR3 is a novel antagonist of the canonical transforming growth factor ß (TGFß) pathway, which induces potent migration and invasion responses in breast cancer cells. METHODS: We surveyed functional genomics databases for correlations between BCAR3 expression and disease outcomes of breast cancer patients. We also studied how BCAR3 could regulate the TGFß/Smad signaling axis using Western blot analysis, coimmunoprecipitation and luciferase assays. In addition, we examined whether BCAR3 could modulate TGFß-induced cell migration and invasion by using an automated imaging system and a confocal microscopy imaging-based matrix degradation assay, respectively. RESULTS: Relatively low levels of BCAR3 expression in primary breast tumors correlate with poor distant metastasis-free survival and relapse-free survival outcomes. We also found a strong correlation between the loss of heterozygosity at BCAR3 gene alleles and lymph node invasion in human breast cancer, further suggesting a role for BCAR3 in preventing disease progression. In addition, we found BCAR3 to inhibit Smad activation, Smad-mediated gene transcription, Smad-dependent cell migration and matrix digestion in breast cancer cells. Furthermore, we found BCAR3 to be downregulated by TGFß through proteasome degradation, thus defining a novel positive feedback loop mechanism downstream of the TGFß/Smad signaling pathway. CONCLUSION: BCAR3 is considered to be associated with aggressive breast cancer phenotypes. However, our results indicate that BCAR3 acts as a putative suppressor of breast cancer progression by inhibiting the prometastatic TGFß/Smad signaling pathway in invasive breast tumors. These data provide new insights into BCAR3's molecular mechanism of action and highlight BCAR3 as a novel TGFß/Smad antagonist in breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adenocarcinoma/genética , Neoplasias de la Mama/genética , ARN Mensajero/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Factores de Intercambio de Guanina Nucleótido , Humanos , Células MCF-7 , Pronóstico , Transducción de Señal , Tamoxifeno/uso terapéutico
7.
Calcif Tissue Int ; 94(4): 454-64, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24362451

RESUMEN

The osteoinductive factors BMP-2 and Tmem119 that promote the differentiation of myoblasts into osteoblasts, each increase the levels of the other. However, the relative contributions of BMP-2 and Tmem119 to the osteogenic differentiation and the mechanisms involved are incompletely understood. In the present study, we examined the relationship among BMP-2, Tmem119, and the PERK-eIF2α-ATF4 endoplasmic reticulum (ER) stress response pathway in the differentiation of C2C12 myoblasts into osteoblastic cells. Both BMP-2 and Tmem119 induced levels of the osteoblast markers Runx2, Osterix, Col1a1, ALP, and osteocalcin, as well as mineralization. BMP-2 activation of the ER stress sensor PERK stimulated phosphorylation of eIF2α and led to increased biosynthesis of the osteoblast differentiation factor ATF4. When dephosphorylation of eIF2α was blocked by the selective inhibitor salubrinal, the osteogenic effects of BMP-2 and Tmem119 were enhanced further. Although BMP-2 stimulated both P-eIF2α and ATF4 levels, Tmem119 had no effect on P-eIF2α but stimulated ATF4 only. Reduction in endogenous Tmem119 levels by siRNA reduced both basal and BMP-2-stimulated levels of the ATF4 protein. In conclusion, BMP-2 stimulates differentiation of myoblasts into osteoblasts via the PERK-eIF2α-ATF4 pathway but in addition stimulates Tmem119, which itself increases ATF4. Hence, BMP-2 stimulates ATF4 both dependently and independently of the PERK-eIF2α ER stress response pathway.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Estrés del Retículo Endoplásmico , Proteínas de la Membrana/metabolismo , Mioblastos/citología , Osteoblastos/citología , Proteínas Serina-Treonina Quinasas/metabolismo , eIF-2 Quinasa/metabolismo , Células 3T3 , Animales , Diferenciación Celular , Línea Celular , Cinamatos/química , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Mioblastos/metabolismo , Osteoblastos/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Tiourea/análogos & derivados , Tiourea/química
8.
Cells ; 13(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38891107

RESUMEN

Over the past few decades, the worldwide incidence of cutaneous melanoma, a malignant neoplasm arising from melanocytes, has been increasing markedly, leading to the highest rate of skin cancer-related deaths. While localized tumors are easily removed by excision surgery, late-stage metastatic melanomas are refractory to treatment and exhibit a poor prognosis. Consequently, unraveling the molecular mechanisms underlying melanoma tumorigenesis and metastasis is crucial for developing novel targeted therapies. We found that the multiple endocrine neoplasia type 1 (MEN1) gene product Menin is required for the transforming growth factor beta (TGFß) signaling pathway to induce cell growth arrest and apoptosis in vitro and prevent tumorigenesis in vivo in preclinical xenograft models of melanoma. We further identified point mutations in two MEN1 family members affected by melanoma that led to proteasomal degradation of the MEN1 gene product and to a loss of TGFß signaling. Interestingly, blocking the proteasome degradation pathway using an FDA-approved drug and RNAi targeting could efficiently restore MEN1 expression and TGFß transcriptional responses. Together, these results provide new potential therapeutic strategies and patient stratification for the treatment of cutaneous melanoma.


Asunto(s)
Melanoma , Transducción de Señal , Factor de Crecimiento Transformador beta , Melanoma/genética , Melanoma/patología , Melanoma/metabolismo , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular Tumoral , Ratones , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Apoptosis/genética , Carcinogénesis/genética , Carcinogénesis/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica
9.
J Biol Chem ; 287(11): 8584-97, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22275377

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is characterized by tumors of the parathyroid, enteropancreas, and anterior pituitary. The MEN1 gene encodes the tumor suppressor menin of 610 amino acids that has multiple protein partners and activities. The particular pathways that, when lost, lead to tumorigenesis are not known. We demonstrated that members of a three-generation MEN1 kindred are heterozygous for a donor splice site mutation at the beginning of intron 3 (IVS3 + 1G→A). Lymphoblastoid cells of a mutant gene carrier had, in addition to the wild-type menin transcript, an aberrant transcript resulting from use of a cryptic splice site within exon III that splices to the start of exon IV. The predicted menin Δ(184-218) mutant has an in-frame deletion of 35 amino acids but is otherwise of wild-type sequence. The transfected menin Δ(184-218) mutant was well expressed and fully able to mediate the normal inhibition of the activity of the transcriptional regulators JunD and NF-κB. However, it was defective in mediating TGF-ß-stimulated Smad3 action in promoter-reporter assays in insulinoma cells. Importantly, lymphoblastoid cells from an individual heterozygous for the mutation had reduced TGF-ß-induced (Smad3) transcriptional activity but normal JunD and NF-κB function. In addition, the mutant gene carrier lymphoblastoid cells proliferated faster and were less responsive to the cytostatic effects of TGF-ß than cells from an unaffected family member. In conclusion, the menin mutant exhibits selective loss of the TGF-ß signaling pathway and loss of cell proliferation control contributing to the development of MEN1.


Asunto(s)
Secuencia de Aminoácidos/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sitios de Empalme de ARN/genética , Eliminación de Secuencia , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adolescente , Adulto , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Intrones , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética
10.
J Biol Chem ; 286(11): 9787-96, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21239498

RESUMEN

The mechanisms whereby the parathyroid hormone (PTH) exerts its anabolic action on bone are incompletely understood. We previously showed that inhibition of ERK1/2 enhanced Smad3-induced bone anabolic action in osteoblasts. These findings suggested the hypothesis that changes in gene expression associated with the altered Smad3-induced signaling brought about by an ERK1/2 inhibitor would identify novel bone anabolic factors in osteoblasts. We therefore performed a comparative DNA microarray analysis between empty vector-transfected mouse osteoblastic MC3T3-E1 cells and PD98059-treated stable Smad3-overexpressing MC3T3-E1 cells. Among the novel factors, Tmem119 was selected on the basis of its rapid induction by PTH independent of later increases in endogenous TGF-ß. The levels of Tmem119 increased with time in cultures of MC3T3-E1 cells and mouse mesenchymal ST-2 cells committed to the osteoblast lineage by BMP-2. PTH stimulated Tmem119 levels within 1 h as determined by Western blot analysis and immunocytochemistry in MC3T3-E1 cells. MC3T3-E1 cells stably overexpressing Tmem119 exhibited elevated levels of Runx2, osteocalcin, alkaline phosphatase, and ß-catenin, whereas Tmem119 augmented BMP-2-induced Runx2 levels in mesenchymal cells. Tmem119 interacted with Runx2, Smad1, and Smad5 in C2C12 cells. In conclusion, we identified a Smad3-related factor, Tmem119, that is induced by PTH and promotes differentiation in mouse osteoblastic cells. Tmem119 is an important molecule in the pathway downstream of PTH and Smad3 signaling in osteoblasts.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de la Membrana/metabolismo , Osteoblastos/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/genética , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Osteoblastos/citología , Osteocalcina/genética , Osteocalcina/metabolismo , Hormona Paratiroidea/farmacología , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo
11.
Endocr J ; 59(8): 653-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22673292

RESUMEN

The transforming growth factor (TGF)-ß family members, bone morphogenetic protein (BMP)-2 and TGF-ß that signal via the receptor-regulated Smads (R-Smads) induce bone formation in vivo. The inhibitory Smads (I-Smads), Smad6 and Smad7, negatively regulate TGF-ß family ligand signaling by competing with R-Smads for binding to activated type I receptors, and preventing R-Smad activation, Hence, the I-Smads potentially act as suppressors of bone formation although their effects on phenotypic changes in mature osteoblasts are unclear. While Smad7 inhibits both BMP and TGF-ß signaling, Smad6 is less effective in inhibiting TGF-ß signaling. The present study was performed to examine the role of Smad7 on the phenotype of mouse osteoblastic MC3T3-E1 cells. We employed stable Smad7-transfected MC3T3-E1 cells to examine the role of Smad7 in osteoblast proliferation, differentiation and mineralization. Stable Smad7 overexpression significantly inhibited the absorbance in the MTT-dye assay and inhibited the levels of PCNA compared with those in empty vector-transfected cells. Smad7 overexpression suppressed the type 1 collagen mRNA and protein levels. Moreover, Smad7 inhibited ALP activity and mineralization of osteoblastic cells. The effects of stable overexpression of Smad6 were similar to those of Smad7 suggesting the changes mediated by either I-Smad occurred by inhibition of BMP rather than TGF-ß signaling. In addition, PTH-(1-34) elevated the levels of Smad7 in parental MC3T3-E1 cells. In conclusion, the present study demonstrated that Smad7, as well as Smad6, inhibits proliferation, differentiation and mineralization of mouse osteoblastic cells. Therefore, I-Smads are important molecular targets for the negative control of bone formation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Proteína smad7/farmacología , Fosfatasa Alcalina/metabolismo , Animales , Colágeno Tipo I/biosíntesis , Ratones , Hormona Paratiroidea/farmacología , ARN Interferente Pequeño/farmacología , Proteína smad6/metabolismo , Proteína smad6/farmacología , Proteína smad7/metabolismo
12.
JBMR Plus ; 6(5): e10622, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35509630

RESUMEN

Loss-of-function mutations in the MEN1 tumor-suppressor gene cause the multiple endocrine neoplasia type 1 syndrome. Menin, the MEN1 gene product, is expressed in many tissues, including bone, where its function remains elusive. We conditionally inactivated menin in mesenchymal stem cells (MSCs) using paired-related homeobox 1 (Prx1)-Cre and compared resultant skeletal phenotypes of Prx1-Cre;Men1 f/f menin-knockout mice (KO) and wild-type controls using in vivo and in vitro experimental approaches and mechanics simulation. Dual-energy X-ray absorptiometry demonstrated significantly reduced bone mineral density, and 3-dimensional micro-CT imaging revealed a decrease in trabecular bone volume, altered trabecular structure, and an increase in trabecular separation in KO mice at 6 and 9 months of age. Numbers of osteoblasts were unaltered, and dynamic histomorphometry demonstrated unaltered bone formation; however, osteoclast number and activity and receptor activator of NF-κB ligand/osteoprotegerin (RANKL/OPG) mRNA profiles were increased, supporting increased osteoclastogenesis and bone resorption. In vitro, proliferative capabilities of bone marrow stem cells and differentiation of osteoblasts and mineralization were unaltered; however, osteoclast generation was increased. Gross femur geometrical alterations observed included significant reductions in length and in mid-metaphyseal cross-sectional area. Atomic force microscopy demonstrated significant decreases in elasticity of both cortical and trabecular bone at the nanoscale, whereas three-point bending tests demonstrated a 30% reduction in bone stiffness; finite element analysis showed morphological changes of the femur microgeometry and a significantly diminished femur flexural rigidity. The biomechanical results demonstrated the detrimental outcome of the accelerated osteoclastic bone resorption. Our studies have a twofold implication; first, MEN1 deletion from MSCs can negatively regulate bone mass and bone biomechanics, and second, the experimental and computational biomechanical analyses employed in the present study should be applicable for improved phenotypic characterization of murine bone. Furthermore, our findings of critical menin function in bone may underpin the more severe skeletal phenotype found in hyperparathyroidism associated with loss-of-function of the MEN1 gene. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

13.
Biomimetics (Basel) ; 7(3)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35892371

RESUMEN

Bone has a complex microenvironment formed by an extracellular matrix (ECM) composed mainly of mineralized type I collagen fibres. Bone ECM regulates signaling pathways important in the differentiation of osteoblast-lineage cells, necessary for bone mineralization and in preserving tissue architecture. Compared to conventional 2D cell cultures, 3D in vitro models may better mimic bone ECM and provide an environment to support osteoblastic differentiation. In this study, a biomimetic 3D osteoid-like dense collagen gel model was used to investigate the role of the nuclear protein menin plays in osteoblastic differentiation and matrix mineralization. Previous in vitro and in vivo studies have shown that when expressed at later stages of osteoblastic differentiation, menin modulates osteoblastogenesis and regulates bone mass in adult mice. To investigate the role of menin when expressed at earlier stages of the osteoblastic lineage, conditional knockout mice in which the Men1 gene is specifically deleted early (i.e., at the level of the pluripotent mesenchymal stem cell lineage), where generated and primary calvarial osteoblasts were cultured in plastically compressed dense collagen gels for 21 days. The proliferation, morphology and differentiation of isolated seeded primary calvarial osteoblasts from knockout (Prx1-Cre; Men1f/f) mice were compared to those isolated from wild-type (Men1f/f) mice. Primary calvarial osteoblasts from knockout and wild-type mice did not show differences in terms of proliferation. However, in comparison to wild-type cells, primary osteoblast cells derived from knockout mice demonstrated deficient mineralization capabilities and an altered gene expression profile when cultured in 3D dense collagen gels. In summary, these findings indicate that when expressed at earlier stages of osteoblast differentiation, menin is important in maintaining matrix mineralization in 3D dense collagen gel matrices, in vitro.

14.
Eur J Endocrinol ; 186(3): 351-366, 2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35038313

RESUMEN

OBJECTIVE: The aim of this study was to analyze variants of the gene glial cells missing-2 (GCM2), encoding a parathyroid cell-specific transcription factor, in familial hypoparathyroidism and in familial isolated hyperparathyroidism (FIHP) without and with parathyroid carcinoma. DESIGN: We characterized 2 families with hypoparathyroidism and 19 with FIHP in which we examined the mechanism of action of GCM2 variants. METHODS: Leukocyte DNA of hypoparathyroid individuals was Sanger sequenced for CASR, PTH, GNA11 and GCM2 mutations. DNA of hyperparathyroid individuals underwent MEN1, CDKN1B, CDC73, CASR, RET and GCM2 sequencing. The actions of identified GCM2 variants were evaluated by in vitro functional analyses. RESULTS: A novel homozygous p.R67C GCM2 mutation which failed to stimulate transcriptional activity in a luciferase assay was identified in affected members of two hypoparathyroid families. Oligonucleotide pull-down assay and in silico structural modeling indicated that this mutant had lost the ability to bind the consensus GCM recognition sequence of DNA. Two novel (p.I383M and p.T386S) and one previously reported (p.Y394S) heterozygous GCM2 variants that lie within a C-terminal conserved inhibitory domain were identified in three affected individuals of the hyperparathyroid families. One family member, heterozygous for p.I138M, had parathyroid carcinoma (PC), and a heterozygous p.V382M variant was found in another patient affected by sporadic PC. These variants exerted significantly enhanced in vitrotranscriptional activity, including increased stimulation of the PTH promoter. CONCLUSIONS: We provide evidence that two novel GCM2 R67C inactivating mutations with an inability to bind DNA are causative of hypoparathyroidism. Additionally, we provide evidence that two novel GCM2 variants increased transactivation of the PTH promoter in vitro and are associated with FIHP. Furthermore, our studies suggest that activating GCM2 variants may contribute to facilitating more aggressive parathyroid disease.


Asunto(s)
Hiperparatiroidismo/genética , Hipoparatiroidismo/genética , Mutación , Proteínas Nucleares/genética , Neoplasias de las Paratiroides/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Sitios de Unión , Calcio/sangre , Calcio/orina , ADN/sangre , ADN/metabolismo , Femenino , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/patología , Hipoparatiroidismo/sangre , Lactante , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Glándulas Paratiroides/patología , Glándulas Paratiroides/cirugía , Hormona Paratiroidea/sangre , Hormona Paratiroidea/genética , Neoplasias de las Paratiroides/metabolismo , Neoplasias de las Paratiroides/patología , Linaje , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Factores de Transcripción/química , Factores de Transcripción/metabolismo
15.
Hum Mutat ; 30(1): 85-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18712808

RESUMEN

Glial cells missing-2 (GCM2) is a transcription factor expressed in the parathyroid hormone (PTH)-secreting cells of the parathyroid gland and is essential for their development. Thus far, downstream targets of GCM2 have not been identified. Here, we show that both promoters (P1 and P2) of the calcium-sensing receptor (CASR) gene, a differentiation marker for the parathyroid gland, are transactivated by wild-type GCM2. GCM response elements within CASR P1 (-451 to -441; relative to the transcription start site at +1) and CASR P2 (-166 to -156) were identified by mutated promoter-reporter studies as well as oligonucleotide precipitation assays. Primary hypoparathyroidism is a heterogeneous group of conditions characterized by hypocalcemia and hyperphosphatemia due to deficient PTH secretion. A few cases of familial isolated hypoparathyroidism with autosomal recessive inheritance have been identified that are caused by homozygous inactivating mutations in the GCM2 gene. We describe the GCM2 mutations in two families with hypoparathyroidism, one inherited in an autosomal recessive fashion and the other in an autosomal dominant manner. In transfection studies using a promoter-reporter construct having synthetic multimerized GCM elements in the promoter, the dominantly inherited mutant GCM2 exerted a dominant-negative effect on wild-type GCM2 activity, whereas recessively inherited mutants did not. In addition, we show that the transactivation of the CASR promoter-reporter constructs by wild-type GCM2 is completely abolished in the presence of the dominant-negative mutant GCM2.


Asunto(s)
Hipoparatiroidismo/genética , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Sensibles al Calcio/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Células Cultivadas , Femenino , Genes Dominantes , Humanos , Recién Nacido , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Linaje , Regiones Promotoras Genéticas , Receptores Sensibles al Calcio/metabolismo , Transcripción Genética
16.
J Cell Biochem ; 108(1): 285-94, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19582775

RESUMEN

Parathyroid hormone (PTH) exerts an anabolic action on bone but the mechanisms are incompletely understood. We showed previously that PTH interacts with the canonical Wnt-beta-catenin signaling pathway via the transforming growth factor (TGF)-beta signaling molecule, Smad3, to modulate osteoblast differentiation and apoptosis. Here, we examined which actions of Smad3 are TGF-beta-independent in stimulating the osteoblast phenotype and PTH-induced Wnt-beta-catenin signaling. For this, the TGF-beta receptor type 1 [activin receptor-like kinase (ALK5)] inhibitor (SB431542), and a Smad3 mutant in which the site normally phosphorylated by ALK5 is mutated from SSVS to AAVA, was used. PTH induced total beta-catenin and reduced phosphorylated beta-catenin levels at 1, 6, and 24 h in mouse osteoblastic MC3T3-E1 cells. Transient transfection of Smad3AAVA inhibited the PTH induction of total beta-catenin and reduction of phosphorylated beta-catenin levels at 6 and 24 h, but not at 1 h, indicating that the early effects occur independently of TGF-beta receptor signaling. On the other hand, MC3T3-E1 cell clones in which Smad3AAVA was stably expressed demonstrated elevated beta-catenin levels, although alkaline phosphatase (ALP) activity and mineralization were unaltered. In contrast, MC3T3-E1 cell clones in which wild-type Smad3 was stably expressed exhibited increased ALP activity and mineralization that were decreased by the ALK5 inhibitor, SB431542, although the beta-catenin levels induced in these cells were not modulated. In conclusion, the present study indicates that PTH induces osteoblast beta-catenin levels via Smad3 independently of, and dependently on, TGF-beta in the early and later induction phases, respectively.


Asunto(s)
Osteoblastos/metabolismo , Hormona Paratiroidea/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular , Humanos , Ratones , Fenotipo , Proteína smad3/genética , Transfección , beta Catenina/genética
17.
Adv Exp Med Biol ; 668: 37-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20175451

RESUMEN

Since its discovery as a novel protein some 10 years ago, many cellular functions of menin have been identified. However, which ones of these relate specifically to menin's role as a tumor suppressor and which ones not remains unclear. Menin is predominantly nuclear and acts as a scaffold protein to regulate gene transcription by coordinating chromatin remodeling. It is implicated in both histone deacetylase and histone methyltransferase activity and, via the latter, regulates the expression of cell cycle kinase inhibitor and homeobox domain genes. TGF-beta family members are key cytostatic molecules and menin is a facilitator of the transcriptional activity of their signaling molecules, the Smads, thereby ensuring appropriate control of cell proliferation and differentiation.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Factores de Transcripción Forkhead , GTP Fosfohidrolasas/metabolismo , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteína de Replicación A/metabolismo , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
18.
Adv Exp Med Biol ; 668: 59-67, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20175453

RESUMEN

Menin function is related to transcriptional regulation and cell cycle control and it physically and functionally interacts with osteotropic transcription factors, such as Smad1/5, Smad3, Runx2 and JunD. Menin promotes the commitment of pluripotent mesenchymal stem cells to the osteoblast lineage, mediated by interactions between menin and the BMP signaling molecules, Smad1/5, or Runx2. On the other hand, in mature osteoblasts the interaction of menin and the TGF-beta/Smad3 pathway counteracts the BMP-2/Smad1/5- and Runx2-induced transcriptional activities leading to inhibition of late stage osteoblast differentiation. Moreover, menin suppresses osteoblast maturation partly by inhibiting the differentiation actions of JunD. In conclusion, menin plays an important role in osteoblastogenesis and osteoblast differentiation.


Asunto(s)
Desarrollo Óseo/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Ratones , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Transducción de Señal/fisiología , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
19.
J Clin Endocrinol Metab ; 103(8): 2879-2888, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29846619

RESUMEN

Context: Autosomal dominant hypocalcemia type 1 (ADH1) is caused by heterozygous activating mutations in the calcium-sensing receptor gene (CASR). Whether polymorphisms that are benign in the heterozygous state pathologically alter receptor function in the homozygous state is unknown. Objective: To identify the genetic defect in an adolescent female with a history of surgery for bilateral cataracts and seizures. The patient has hypocalcemia, hyperphosphatemia, and low serum PTH level. The parents of the proband are healthy. Methods: Mutation testing of PTH, GNA11, GCM2, and CASR was done on leukocyte DNA of the proband. Functional analysis in transfected cells was conducted on the gene variant identified. Public single nucleotide polymorphism (SNP) databases were searched for the presence of the variant allele. Results: No mutations were identified in PTH, GNA11, and GCM2 in the proband. However, a germline homozygous variant (c.1631G>A; p.R544Q) in exon 6 of the CASR was identified. Both parents are heterozygous for the variant. The variant allele frequency was near 0.1% in SNP databases. By in vitro functional analysis, the variant was significantly more potent in stimulating both the Ca2+i and MAPK signaling pathways than wild type when transfected alone (P < 0.05) but not when transfected together with wild type. The overactivity of the mutant CaSR is due to loss of a critical structural cation-π interaction. Conclusions: The patient's hypoparathyroidism is due to homozygosity of a variant in the CASR that normally has weak or no phenotypic expression in heterozygosity. Although rare, this has important implications for genetic counseling and clinical management.


Asunto(s)
Hipocalcemia/genética , Hipoparatiroidismo/genética , Polimorfismo de Nucleótido Simple , Receptores Sensibles al Calcio/genética , Sustitución de Aminoácidos , Arginina/genética , Femenino , Ácido Glutámico/genética , Homocigoto , Humanos , Hipocalcemia/complicaciones , Hipoparatiroidismo/complicaciones , Adulto Joven
20.
J Clin Endocrinol Metab ; 92(7): 2616-23, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17473068

RESUMEN

CONTEXT: Familial hypocalciuric hypercalcemia (FHH) is a benign condition associated with heterogeneous inactivating mutations in the calcium-sensing receptor (CASR) gene. OBJECTIVE: The objective of the study was to identify and characterize a CASR mutation in a moderately hypercalcemic, hyperparathyroid individual and his family and assess the influence of vitamin D status on the clinical expression of the defect. SUBJECTS: We studied a kindred with FHH, in which the proband (a 34-yr-old male) was initially diagnosed with primary hyperparathyroidism due to frankly elevated serum PTH levels. METHODS: CASR gene mutation analysis was performed on genomic DNA of the proband and family members. The mutant CASR was functionally characterized by transient transfection studies in kidney cells in vitro. RESULTS: A novel heterozygous mutation (F180C, TTC>TGC) in exon 4 of the CASR gene was identified. Although the mutant receptor was expressed normally at the cell surface, it was unresponsive with respect to intracellular signaling (MAPK activation) to increases in extracellular calcium concentrations. The baby daughter of the proband presented with neonatal hyperparathyroidism with markedly elevated PTH. Vitamin D supplementation of both the proband and the baby resulted in reduction of serum PTH levels to the normal range. The serum calcium level remained at a constant and moderately elevated level. CONCLUSION: The identification of a novel CASR gene mutation established the basis of the hypercalcemia in the kindred. Concomitant vitamin D deficiency modulates the severity of the presentation of FHH.


Asunto(s)
Hipocalcemia/genética , Receptores Sensibles al Calcio/genética , Deficiencia de Vitamina D/genética , Vitamina D/sangre , Adulto , Calcio/sangre , Calcio/orina , Línea Celular , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Heterocigoto , Humanos , Hiperparatiroidismo/genética , Hiperparatiroidismo/metabolismo , Hipocalcemia/metabolismo , Riñón/citología , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Hormona Paratiroidea/sangre , Linaje , Periodo Posparto , Índice de Severidad de la Enfermedad , Vitamina D/administración & dosificación , Deficiencia de Vitamina D/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA