Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nature ; 472(7341): 105-9, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21475200

RESUMEN

Generation of a diverse and self-tolerant T-cell repertoire requires appropriate interpretation of T-cell antigen receptor (TCR) signals by CD4(+ ) CD8(+) double-positive thymocytes. Thymocyte cell fate is dictated by the nature of TCR-major-histocompatibility-complex (MHC)-peptide interactions, with signals of higher strength leading to death (negative selection) and signals of intermediate strength leading to differentiation (positive selection). Molecules that regulate T-cell development by modulating TCR signal strength have been described but components that specifically define the boundaries between positive and negative selection remain unknown. Here we show in mice that repression of TCR-induced death pathways is critical for proper interpretation of positive selecting signals in vivo, and identify schnurri-2 (Shn2; also known as Hivep2) as a crucial death dampener. Our results indicate that Shn2(-/-) double-positive thymocytes inappropriately undergo negative selection in response to positive selecting signals, thus leading to disrupted T-cell development. Shn2(-/-) double-positive thymocytes are more sensitive to TCR-induced death in vitro and die in response to positive selection interactions in vivo. However, Shn2-deficient thymocytes can be positively selected when TCR-induced death is genetically ablated. Shn2 levels increase after TCR stimulation, indicating that integration of multiple TCR-MHC-peptide interactions may fine-tune the death threshold. Mechanistically, Shn2 functions downstream of TCR proximal signalling compenents to dampen Bax activation and the mitochondrial death pathway. Our findings uncover a critical regulator of T-cell development that controls the balance between death and differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Linfocitos T/citología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Muerte Celular , Diferenciación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timo/citología , Timo/inmunología
2.
Proc Natl Acad Sci U S A ; 109(21): 8173-8, 2012 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-22573816

RESUMEN

Mice lacking the large zinc finger protein Schnurri-3 (Shn3) display increased bone mass, in part, attributable to augmented osteoblastic bone formation. Here, we show that in addition to regulating bone formation, Shn3 indirectly controls bone resorption by osteoclasts in vivo. Although Shn3 plays no cell-intrinsic role in osteoclasts, Shn3-deficient animals show decreased serum markers of bone turnover. Mesenchymal cells lacking Shn3 are defective in promoting osteoclastogenesis in response to selective stimuli, likely attributable to reduced expression of the key osteoclastogenic factor receptor activator of nuclear factor-κB ligand. The bone phenotype of Shn3-deficient mice becomes more pronounced with age, and mice lacking Shn3 are completely resistant to disuse osteopenia, a process that requires functional osteoclasts. Finally, selective deletion of Shn3 in the mesenchymal lineage recapitulates the high bone mass phenotype of global Shn3 KO mice, including reduced osteoclastic bone catabolism in vivo, indicating that Shn3 expression in mesenchymal cells directly controls osteoblastic bone formation and indirectly regulates osteoclastic bone resorption.


Asunto(s)
Resorción Ósea/fisiopatología , Proteínas de Unión al ADN/genética , Hiperparatiroidismo Secundario/fisiopatología , Osteoblastos/fisiología , Osteoclastos/fisiología , Envejecimiento/fisiología , Animales , Resorción Ósea/genética , Células Cultivadas , Técnicas de Cocultivo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Hiperparatiroidismo Secundario/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Fenotipo , Ligando RANK/metabolismo , Elementos Reguladores de la Transcripción/fisiología , Cráneo/citología
3.
Nat Rev Immunol ; 2(10): 748-59, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12360213

RESUMEN

Lipids and lipid metabolism have well-documented regulatory effects on inflammatory processes. Recent work has highlighted the role of the peroxisome proliferator-activated receptors (PPARs)--a subset of the nuclear-hormone-receptor superfamily that are activated by various lipid species--in regulating inflammatory responses. Here, we describe how the PPARs, through their interactions with transcription factors and other cell-signalling systems, have important regulatory roles in innate and adaptive immunity.


Asunto(s)
Inflamación/etiología , Receptores Citoplasmáticos y Nucleares/inmunología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/inmunología , Factores de Transcripción/fisiología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunidad Innata , Inflamación/tratamiento farmacológico , Ligandos , Modelos Inmunológicos , Receptores Citoplasmáticos y Nucleares/química , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Transcripción/química , Activación Transcripcional
4.
Proc Natl Acad Sci U S A ; 107(18): 8254-8, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404140

RESUMEN

Formation and remodeling of the skeleton relies on precise temporal and spatial regulation of genes expressed in cartilage and bone cells. Debilitating diseases of the skeletal system occur when mutations arise that disrupt these intricate genetic regulatory programs. Here, we report that mice bearing parallel null mutations in the adapter proteins Schnurri2 (Shn2) and Schnurri3 (Shn3) exhibit defects in patterning of the axial skeleton during embryogenesis. Postnatally, these compound mutant mice develop a unique osteochondrodysplasia. The deletion of Shn2 and Shn3 impairs growth plate maturation during endochondral ossification but simultaneously results in massively elevated trabecular bone formation. Hence, growth plate maturation and bone formation can be uncoupled under certain circumstances. These unexpected findings demonstrate that both unique and redundant functions reside in the Schnurri protein family that are required for proper skeletal patterning and remodeling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/metabolismo , Osteogénesis , Animales , Densidad Ósea , Proteínas de Unión al ADN/deficiencia , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Placa de Crecimiento/embriología , Ratones , Ratones Noqueados , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Fenotipo
5.
J Clin Invest ; 118(2): 491-504, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18219387

RESUMEN

Drug targeting of adult stem cells has been proposed as a strategy for regenerative medicine, but very few drugs are known to target stem cell populations in vivo. Mesenchymal stem/progenitor cells (MSCs) are a multipotent population of cells that can differentiate into muscle, bone, fat, and other cell types in context-specific manners. Bortezomib (Bzb) is a clinically available proteasome inhibitor used in the treatment of multiple myeloma. Here, we show that Bzb induces MSCs to preferentially undergo osteoblastic differentiation, in part by modulation of the bone-specifying transcription factor runt-related transcription factor 2 (Runx-2) in mice. Mice implanted with MSCs showed increased ectopic ossicle and bone formation when recipients received low doses of Bzb. Furthermore, this treatment increased bone formation and rescued bone loss in a mouse model of osteoporosis. Thus, we show that a tissue-resident adult stem cell population in vivo can be pharmacologically modified to promote a regenerative function in adult animals.


Asunto(s)
Regeneración Ósea , Ácidos Borónicos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/citología , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Animales , Bortezomib , Diferenciación Celular , Femenino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL
6.
Adv Exp Med Biol ; 658: 117-22, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19950022

RESUMEN

Although identified over a decade ago, the function and physiological significance of the mammalian Schnurri protein family remained largely unknown. However, the recent generation and characterization of mice bearing null mutations in the individual Schnurri genes has led to the discovery of unexpected yet central roles for these large zinc-finger proteins in several biological processes. Here, we review findings of these studies and discuss the importance of the Schnurri protein family in regulating both the immune and skeletal systems.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Osteogénesis , Animales , Huesos/metabolismo , Humanos
7.
JBMR Plus ; 3(11): e10226, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31768487

RESUMEN

Schnurri-3 (Shn3) is an essential regulator of postnatal skeletal remodeling. Shn3-deficient mice (Shn3-/-) have high bone mass; however, their bone mechanical and material properties have not been investigated to date. We performed three-point bending of femora, compression tests of L3 vertebrae. We also measured intrinsic material properties, including bone mineralization density distribution (BMDD) and osteocyte lacunae section (OLS) characteristics by quantitative backscatter electron imaging, as well as collagen cross-linking by Fourier transform infrared microspectroscopy of femora from Shn3-/- and WT mice at different ages (6 weeks, 4 months, and 18 months). Moreover, computer modeling was performed for the interpretation of the BMDD outcomes. Femora and L3 vertebrae from Shn3-/- aged 6 weeks revealed increased ultimate force (2.2- and 3.2-fold, p < .01, respectively). Mineralized bone volume at the distal femoral metaphysis was about twofold (at 6 weeks) to eightfold (at 4 and 18 months of age) in Shn3-/- (p < .001). Compared with WT, the average degree of trabecular bone mineralization was similar at 6 weeks, but increased at 4 and 18 months of age (+12.6% and +7.7%, p < .01, respectively) in Shn3-/-. The analysis of OLS characteristics revealed a higher OLS area for Shn3-/- versus WT at all ages (+16%, +23%, +21%, respectively, p < .01). The collagen cross-link ratio was similar between groups. We conclude that femora and vertebrae from Shn3-/- had higher ultimate force in mechanical testing. Computer modeling demonstrated that in cases of highly increased bone volume, the average degree of bone matrix mineralization can be higher than in WT bone, which was actually measured in the older Shn3-/- groups. The area of 2D osteocyte lacunae sections was also increased in Shn3-deficiency, which could only partly be explained by larger remnant areas of primary cortical bone. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

8.
Methods Mol Biol ; 455: 149-55, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18463817

RESUMEN

Osteoblasts are the sole cell responsible for bone formation in vivo (1). Although genetic techniques have been extremely valuable to study the functions of certain genes in these cells in vivo, this approach is time consuming and expensive. An alternative loss-of-function approach that has been validated in many mammalian systems is shRNA-mediated gene silencing. This chapter describes methodology designed to introduce shRNA constructs into primary murine osteoblasts ex vivo in order to quickly assess the function of genes in osteoblast differentiation and extra cellular matrix mineralization. Both the production of shRNA-expressing lentiviruses and the infection of calvarial osteoblasts with these lentiviruses are detailed.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus , Osteoblastos/fisiología , ARN/metabolismo , Animales , Células Cultivadas , Silenciador del Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Células 3T3 NIH , Osteoblastos/citología , ARN/genética , Cráneo/citología
9.
Ann N Y Acad Sci ; 1116: 174-81, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18083927

RESUMEN

The completed skeleton undergoes continuous remodeling for the duration of adult life. Rates of bone formation by osteoblasts and bone resorption by osteoclasts determine adult bone mass. Abnormalities in either the osteoblast or osteoclast compartment affect bone mass and result in skeletal disorders, the most common of which is osteoporosis, a state of low bone mass. Much is known about the molecular control of bone formation and resorption from rare single gene disorders resulting in elevated or reduced bone mass. Such genetic disorders can be attributed either to osteoclast deficiencies, collectively termed "osteopetrosis," or to intrinsically elevated osteoblast activity, termed "osteosclerosis." However, an increasing need for anabolic therapies to prevent age-induced bone loss has stimulated a search for additional genes that act at the level of the osteoblast to regulate matrix synthesis. Recently, we have discovered a zinc finger adaptor protein called Schnurri-3 (Shn3) that potently regulates adult bone mass. Mice that lack Shn3 have normal skeletal morphogenesis but display profoundly elevated bone mass that increases with age. The molecular mechanism was revealed to be the recruitment of WWP1, a Nedd4 family E3 ubiquitin ligase, by Shn3 to the major transcriptional regulator of the osteoblast, Runx2. In the absence of Shn3, Runx2 degradation by WWP1 is inhibited resulting in increased levels of Runx2 protein and enhanced expression of Runx2 target genes leading to increased osteoblast synthetic activity. Small molecules that inhibit Shn3 or WWP1 may be attractive candidates for the treatment of diseases of low bone mass.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Tamaño de los Órganos/fisiología , Osteogénesis/fisiología , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Ratones , Ubiquitina-Proteína Ligasas/fisiología
10.
Adv Exp Med Biol ; 602: 1-13, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17966382

RESUMEN

Schnurri-3, a large zinc finger protein distantly related to Drosophila Shn, is a potent and essential regulator of adult bone formation. Mice lacking Shn3 display an osteosclerotic phenotype with profoundly increased bone mass due to augmented osteoblast activity. Shn3 controls protein levels of Runx2, the principal regulator of osteoblast differentiation, by promoting its degradation. In osteoblasts, Shn3 functions as a component of a trimeric complex between Runx2 and the E3 ubiquitin ligase WWP1. This complex inhibits Runx2 function and expression of genes involved in extracellular matrix mineralization due to the ability of WWP1 to promote Runx2 polyubiquitination and proteasome-dependent degradation. Our study reveals an essential role for Shn3 as a regulator of postnatal bone mass. Compounds designed to block Shn3/WWP1 function may be possible therapeutic agents for the treatment of osteoporosis.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Densidad Ósea , Huesos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ratones , Osteoblastos/citología , Osteoclastos/metabolismo , Osteoporosis/fisiopatología , Ubiquitina-Proteína Ligasas/genética
11.
PLoS One ; 12(7): e0180870, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28719615

RESUMEN

While the immune system is essential for the maintenance of the homeostasis, health and survival of humans, aberrant immune responses can lead to chronic inflammatory and autoimmune disorders. Pharmacological modulation of drug targets in the immune system to ameliorate disease also carry a risk of immunosuppression that could lead to adverse outcomes. Therefore, it is important to understand the 'immune fingerprint' of novel therapeutics as they relate to current and, clinically used immunological therapies to better understand their potential therapeutic benefit as well as immunosuppressive ability that might lead to adverse events such as infection risks and cancer. Since the mechanistic investigation of pharmacological modulators in a drug discovery setting is largely compound- and mechanism-centric but not comprehensive in terms of immune system impact, we developed a human tissue based functional assay platform to evaluate the impact of pharmacological modulators on a range of innate and adaptive immune functions. Here, we demonstrate that it is possible to generate a qualitative and quantitative immune system impact of pharmacological modulators, which might help better understand and predict the benefit-risk profiles of these compounds in the treatment of immune disorders.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Sistema Inmunológico/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Quimiocinas/biosíntesis , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Fagocitos/efectos de los fármacos , Fagocitos/inmunología , Fagocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Receptores Toll-Like/metabolismo , Transcriptoma/efectos de los fármacos
12.
Front Plant Sci ; 7: 983, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27462324

RESUMEN

Nearly immobile, plants have evolved new components to be able to respond to changing environments. One example is Qua Quine Starch (QQS, AT3G30720), an Arabidopsis thaliana-specific orphan gene that integrates primary metabolism with adaptation to environment changes. SAQR (Senescence-Associated and QQS-Related, AT1G64360), is unique to a clade within the family Brassicaceae; as such, the gene may have arisen about 20 million years ago. SAQR is up-regulated in QQS RNAi mutant and in the apx1 mutant under light-induced oxidative stress. SAQR plays a role in carbon allocation: overexpression lines of SAQR have significantly decreased starch content; conversely, in a saqr T-DNA knockout (KO) line, starch accumulation is increased. Meta-analysis of public microarray data indicates that SAQR expression is correlated with expression of a subset of genes involved in senescence, defense, and stress responses. SAQR promoter::GUS expression analysis reveals that SAQR expression increases after leaf expansion and photosynthetic capacity have peaked, just prior to visible natural senescence. SAQR is expressed predominantly within leaf and cotyledon vasculature, increasing in intensity as natural senescence continues, and then decreasing prior to death. In contrast, under experimentally induced senescence, SAQR expression increases in vasculature of cotyledons but not in true leaves. In SAQR KO line, the transcript level of the dirigent-like disease resistance gene (AT1G22900) is increased, while that of the Early Light Induced Protein 1 gene (ELIP1, AT3G22840) is decreased. Taken together, these data indicate that SAQR may function in the QQS network, playing a role in integration of primary metabolism with adaptation to internal and environmental changes, specifically those that affect the process of senescence.

13.
Cell Rep ; 17(12): 3206-3218, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009290

RESUMEN

Recent studies have elucidated the molecular mechanism of RORγT transcriptional regulation of Th17 differentiation and function. RORγT was initially identified as a transcription factor required for thymopoiesis by maintaining survival of CD4+CD8+ (DP) thymocytes. While RORγ antagonists are currently being developed to treat autoimmunity, it remains unclear how RORγT inhibition may impact thymocyte development. In this study, we show that in addition to regulating DP thymocytes survival, RORγT also controls genes that regulate thymocyte migration, proliferation, and T cell receptor (TCR)α selection. Strikingly, pharmacological inhibition of RORγ skews TCRα gene rearrangement, limits T cell repertoire diversity, and inhibits development of autoimmune encephalomyelitis. Thus, targeting RORγT not only inhibits Th17 cell development and function but also fundamentally alters thymic-emigrant recognition of self and foreign antigens. The analysis of RORγ inhibitors has allowed us to gain a broader perspective of the diverse function of RORγT and its impact on T cell biology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timocitos/inmunología , Animales , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/terapia , Regulación de la Expresión Génica/inmunología , Reordenamiento Génico/genética , Humanos , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Células Th17/efectos de los fármacos , Células Th17/inmunología
14.
Antioxid Redox Signal ; 5(5): 537-48, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14580308

RESUMEN

The elderly suffer impairments to their immune system, evidenced by higher susceptibility to infections, cancer, and many diseases believed to be autoimmune in nature. A dysregulated overexpression of many proinflammatory cytokines also occurs with aging, as does the synthesis of enzymes that control expression of inflammatory lipid mediators and reactive oxygen species. An inappropriate activation of redox-controlled transcription factors, like nuclear factor-kappaB, occurs in many tissues from aged donors, and has been linked to excesses in cellular oxidative stress. Recently, the peroxisome proliferator-activated receptor-alpha (PPARalpha) has been evaluated for its effects on inflammatory and adaptive immune processes. PPARalpha provides redox-balancing influences on various lymphoid cell types and their inducible responses. We recently discovered that PPARalpha transiently suppresses the transcription of gamma-interferon (IFNgamma) by inhibiting the induction of T-bet. We now report that PPARalpha expression in CD4+ T cells is affected by the aging process. Lower PPARalpha levels are present in aged CD4+ T cells, and appear responsible for the suppressed interleukin-2 and exaggerated IFNgamma responses by these cells. Restoration of PPARalpha, T-bet, interleukin-2, and IFNgamma responses was found in T cells from aged animals supplemented with vitamin E, suggesting that interventions that focus on restoring redox balance might benefit the ailing aged immune system.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/fisiología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Administración Oral , Factores de Edad , Animales , Anticuerpos/farmacología , Western Blotting , Antígenos CD28/inmunología , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Expresión Génica , Selectina L/análisis , Selectina L/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Oxidación-Reducción , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Proteínas de Dominio T Box , Factores de Transcripción/genética , Vitamina E/farmacología
15.
J Exp Med ; 210(9): 1793-806, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23918955

RESUMEN

Osteoblasts are responsible for the formation and mineralization of the skeleton. To identify novel regulators of osteoblast differentiation, we conducted an unbiased forward genetic screen using a lentiviral-based shRNA library. This functional genomics analysis led to the identification of the microtubule-associated protein DCAMKL1 (Doublecortin-like and CAM kinase-like 1) as a novel regulator of osteogenesis. Mice with a targeted disruption of Dcamkl1 displayed elevated bone mass secondary to increased bone formation by osteoblasts. Molecular experiments demonstrated that DCAMKL1 represses osteoblast activation by antagonizing Runx2, the master transcription factor in osteoblasts. Key elements of the cleidocranial dysplasia phenotype observed in Runx2(+/-) mice are reversed by the introduction of a Dcamkl1-null allele. Our results establish a genetic linkage between these two proteins in vivo and demonstrate that DCAMKL1 is a physiologically relevant regulator of anabolic bone formation.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microtúbulos/metabolismo , Osteoblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Huesos/metabolismo , Diferenciación Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Quinasas Similares a Doblecortina , Epistasis Genética , Silenciador del Gen , Genómica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lentivirus/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Tamaño de los Órganos , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Fosforilación , Fosfoserina/metabolismo , Polimerizacion , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/metabolismo
16.
J Clin Invest ; 123(9): 4010-22, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23945236

RESUMEN

Mice deficient in Schnurri-3 (SHN3; also known as HIVEP3) display increased bone formation, but harnessing this observation for therapeutic benefit requires an improved understanding of how SHN3 functions in osteoblasts. Here we identified SHN3 as a dampener of ERK activity that functions in part downstream of WNT signaling in osteoblasts. A D-domain motif within SHN3 mediated the interaction with and inhibition of ERK activity and osteoblast differentiation, and knockin of a mutation in Shn3 that abolishes this interaction resulted in aberrant ERK activation and consequent osteoblast hyperactivity in vivo. Additionally, in vivo genetic interaction studies demonstrated that crossing to Lrp5(-/-) mice partially rescued the osteosclerotic phenotype of Shn3(-/-) mice; mechanistically, this corresponded to the ability of SHN3 to inhibit ERK-mediated suppression of GSK3ß. Inducible knockdown of Shn3 in adult mice resulted in a high-bone mass phenotype, providing evidence that transient blockade of these pathways in adults holds promise as a therapy for osteoporosis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Osteoblastos/metabolismo , Vía de Señalización Wnt , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Huesos/metabolismo , Huesos/patología , Huesos/fisiopatología , Proteínas de Unión al ADN/química , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Humanos , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Osteogénesis , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Estructura Terciaria de Proteína , beta Catenina/metabolismo
17.
Ann Rheum Dis ; 66 Suppl 3: iii49-51, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17934096

RESUMEN

Skeletal remodelling is a cyclical process where under normal physiological conditions, bone formation occurs at sites where bone resorption has previously taken place. Homeostatic remodelling of the skeleton is mediated by osteoclasts, giant multinucleated cells of haematopoietic origin that are responsible for bone resorption and osteoblasts, which originate from mesenchymal stem cells, and synthesise the matrix constituents on bone-forming surfaces.1 Proliferation, differentiation and bone remodelling activities of these cells involve a complex temporal network of growth factors, signalling proteins and transcription factors. Dysregulation of any one component may disrupt the remodelling process and contribute to the pathogenesis of common skeletal disorders, like osteoporosis and Paget's disease. Rare single gene disorders resulting in elevated bone mass due to osteoclast defects are collectively termed osteopetrosis. Rarer still are single gene disorders, collectively termed osteosclerosis, in which elevated bone mass is due to intrinsically elevated osteoblast activity.2 While we have learned much about the molecular control of skeletal formation and remodelling from these mutations, additional genes that regulate bone mass have yet to be characterised.


Asunto(s)
Remodelación Ósea/genética , Proteínas de Unión al ADN/genética , Osteoblastos/fisiología , Adulto , Animales , Proteínas de Unión al ADN/antagonistas & inhibidores , Regulación de la Expresión Génica/genética , Humanos , Células Madre Mesenquimatosas/fisiología , Ratones , Persona de Mediana Edad , Osteogénesis/fisiología , Osteoporosis/tratamiento farmacológico , Ubiquitina-Proteína Ligasas/genética , Dedos de Zinc/genética
18.
Science ; 312(5777): 1223-7, 2006 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-16728642

RESUMEN

Genetic mutations that disrupt osteoblast function can result in skeletal dysmorphogenesis or, more rarely, in increased postnatal bone formation. Here we show that Schnurri-3 (Shn3), a mammalian homolog of the Drosophila zinc finger adapter protein Shn, is an essential regulator of adult bone formation. Mice lacking Shn3 display adult-onset osteosclerosis with increased bone mass due to augmented osteoblast activity. Shn3 was found to control protein levels of Runx2, the principal transcriptional regulator of osteoblast differentiation, by promoting its degradation through recruitment of the E3 ubiquitin ligase WWP1 to Runx2. By this means, Runx2-mediated extracellular matrix mineralization was antagonized, revealing an essential role for Shn3 as a central regulator of postnatal bone mass.


Asunto(s)
Densidad Ósea , Huesos/anatomía & histología , Proteínas de Unión al ADN/metabolismo , Animales , Sitios de Unión , Huesos/química , Huesos/fisiología , Línea Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Inmunoprecipitación , Ratones , Osteoblastos/química , Osteoblastos/fisiología , Osteoclastos/fisiología , Osteogénesis , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Activación Transcripcional , Transfección , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Dedos de Zinc
19.
Immunol Rev ; 208: 66-79, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16313341

RESUMEN

Major advances have been made in recent years toward the identification of transcription factors that control cell-type-specific gene expression in the skeletal and adaptive immune systems. However, the identification of factors necessary and sufficient to drive production of effector cell proteins such as matrix components and cytokines represents the first step toward understanding how cells in bone and the adaptive system achieve their highly specialized functions. Here, we provide selected examples of counter-regulatory mechanisms that serve to turn down cells involved in extracellular matrix biosynthesis and adaptive immunity at the level of the transcription factors Runx2 and nuclear factor for the activation of T cells.


Asunto(s)
Huesos/fisiología , Inmunidad , Linfocitos/fisiología , Animales , Matriz Ósea/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Histona Desacetilasas/fisiología , Humanos , Factores de Transcripción NFATC/fisiología , Osteogénesis , PPAR gamma/fisiología , Proteínas Represoras/fisiología , Proteína 1 Relacionada con Twist/fisiología
20.
J Biol Chem ; 277(9): 6838-45, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11726654

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are transcription factors that belong to the nuclear hormone receptor superfamily. PPARalpha and PPARgamma ligands have been demonstrated to exert anti-inflammatory activities in macrophages by repressing the activities of several transcription factors. PPARgamma is expressed in T lymphocytes and may play a role in cytokine production, cellular proliferation, and susceptibility to apoptosis. Herein, we demonstrate that T and B lymphocytes constitutively express PPARalpha. PPARalpha represents the predominant isoform expressed in lymphocytes, whereas PPARgamma dominates in all cell types of the myeloid lineage. PPARalpha expression was down-regulated following T-cell activation while PPARgamma expression increased under the same activating conditions. PPARalpha expression in T cells may be regulated by microenvironmental factors, because Peyer's patch T cells expressed far greater levels of PPARalpha than T cells isolated from peripheral lymphoid organs. Exposure to specific ligand determined that PPARalpha in lymphocytes can effectively transactivate a peroxisome proliferator response element reporter construct. PPARalpha's ability to regulate endogenous genes, however, required treatment with histone deacetylase inhibitors. Finally, ligand activation of lymphocyte PPARalpha antagonized NF-kappaB. Our observation that a functional PPARalpha exists within T cells and B lymphocytes suggests an expanding role for this nuclear receptor in cells of the immune system.


Asunto(s)
Núcleo Celular/metabolismo , Linfocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Animales , Linfocitos B/metabolismo , Butiratos/farmacología , División Celular , Línea Celular , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Femenino , Genes Reporteros , Histona Desacetilasas/metabolismo , Humanos , Interferón gamma/biosíntesis , Ligandos , Luciferasas/metabolismo , Linfocitos/enzimología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Compuestos de Fenilurea/farmacología , Unión Proteica , Isoformas de Proteínas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Factores de Tiempo , Distribución Tisular , Transcripción Genética , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA