Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 574(7777): 249-253, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31578523

RESUMEN

The integrity of the mammalian epidermis depends on a balance of proliferation and differentiation in the resident population of stem cells1. The kinase RIPK4 and the transcription factor IRF6 are mutated in severe developmental syndromes in humans, and mice lacking these genes display epidermal hyperproliferation and soft-tissue fusions that result in neonatal lethality2-5. Our understanding of how these genes control epidermal differentiation is incomplete. Here we show that the role of RIPK4 in mouse development requires its kinase activity; that RIPK4 and IRF6 expressed in the epidermis regulate the same biological processes; and that the phosphorylation of IRF6 at Ser413 and Ser424 primes IRF6 for activation. Using RNA sequencing (RNA-seq), histone chromatin immunoprecipitation followed by sequencing (ChIP-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) of skin in wild-type and IRF6-deficient mouse embryos, we define the transcriptional programs that are regulated by IRF6 during epidermal differentiation. IRF6 was enriched at bivalent promoters, and IRF6 deficiency caused defective expression of genes that are involved in the metabolism of lipids and the formation of tight junctions. Accordingly, the lipid composition of the stratum corneum of Irf6-/- skin was abnormal, culminating in a severe defect in the function of the epidermal barrier. Collectively, our results explain how RIPK4 and IRF6 function to ensure the integrity of the epidermis and provide mechanistic insights into why developmental syndromes that are characterized by orofacial, skin and genital abnormalities result when this axis goes awry.


Asunto(s)
Diferenciación Celular , Células Epidérmicas/citología , Epidermis/fisiología , Factores Reguladores del Interferón/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Anomalías Múltiples/genética , Animales , Labio Leporino/genética , Fisura del Paladar/genética , Quistes/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Células Epidérmicas/metabolismo , Epidermis/embriología , Anomalías del Ojo/genética , Femenino , Dedos/anomalías , Regulación de la Expresión Génica , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Rodilla/anomalías , Articulación de la Rodilla/anomalías , Labio/anomalías , Metabolismo de los Lípidos/genética , Deformidades Congénitas de las Extremidades Inferiores/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sindactilia/genética , Anomalías Urogenitales/genética
2.
Nature ; 559(7712): 120-124, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29950720

RESUMEN

OTULIN (OTU deubiquitinase with linear linkage specificity) removes linear polyubiquitin from proteins that have been modified by LUBAC (linear ubiquitin chain assembly complex) and is critical for preventing auto-inflammatory disease1,2 and embryonic lethality during mouse development3. Here we show that OTULIN promotes rather than counteracts LUBAC activity by preventing its auto-ubiquitination with linear polyubiquitin. Thus, knock-in mice that express catalytically inactive OTULIN, either constitutively or selectively in endothelial cells, resembled LUBAC-deficient mice4 and died midgestation as a result of cell death mediated by TNFR1 (tumour necrosis factor receptor 1) and the kinase activity of RIPK1 (receptor-interacting protein kinase 1). Inactivation of OTULIN in adult mice also caused pro-inflammatory cell death. Accordingly, embryonic lethality and adult auto-inflammation were prevented by the combined loss of cell death mediators: caspase 8 for apoptosis and RIPK3 for necroptosis. Unexpectedly, OTULIN mutant mice that lacked caspase 8 and RIPK3 died in the perinatal period, exhibiting enhanced production of type I interferon that was dependent on RIPK1. Collectively, our results indicate that OTULIN and LUBAC function in a linear pathway, and highlight a previously unrecognized interaction between linear ubiquitination, regulators of cell death, and induction of type I interferon.


Asunto(s)
Muerte Celular , Enzimas Desubicuitinizantes/metabolismo , Endopeptidasas/metabolismo , Inflamación/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinación , Animales , Caspasa 8/genética , Caspasa 8/metabolismo , Muerte Celular/genética , Enzimas Desubicuitinizantes/genética , Pérdida del Embrión/genética , Endopeptidasas/genética , Inflamación/enzimología , Inflamación/genética , Interferón Tipo I/biosíntesis , Ratones , Ratones Endogámicos C57BL , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitinación/genética , Pérdida de Peso/genética
3.
Bioorg Chem ; 116: 105360, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34562676

RESUMEN

Proper recognition of invading pathogens and prompt initiation of host defense mechanisms are instrumental for the maintenance of organismal homeostasis. Nucleotide-binding oligomerization domain-containing (NOD)-like receptors (NLRs) serve as pathogen-recognition receptors that specifically recognize bacterial peptidoglycans. NOD2 detects muramyl dipeptide (MDP) through its carboxy-terminal leucine rich repeats (LRRs), which enables the activation of downstream inflammatory signaling. Synthesis of MDP conjugates based on solution phase chemistry have been previously reported. Our solid phase approach synthetically provides a facile approach for the conjugation of biological probes to MDP, with the advantage of minimal functional/protecting group manipulation, and reduction in the laborious process of intermediate purification and isolation. MDP conjugates that we generated using solid phase synthesis allow detection of NOD2 is cell lysates and NOD2 subcellular localization by immunofluorescence microscopy. MDP-PEG6-Cyanine5.5 conjugate selectively colocalized with WT NOD2 but not NOD2 variant found in Crohn's disease, which lacks carboxy-terminal end and cannot bind MDP. Overall, these data indicate that distinct solid phase-produced MDP conjugates can be used to examine biological properties of NOD2 and could potentially facilitate further development of NOD2 targeting agents.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/síntesis química , Proteína Adaptadora de Señalización NOD2/análisis , Técnicas de Síntesis en Fase Sólida , Células A549 , Acetilmuramil-Alanil-Isoglutamina/química , Células HEK293 , Humanos , Microscopía Fluorescente , Estructura Molecular
4.
Nat Chem Biol ; 14(6): 582-590, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29632413

RESUMEN

Regeneration of the adult intestinal epithelium is mediated by a pool of cycling stem cells, which are located at the base of the crypt, that express leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5). The Frizzled (FZD) 7 receptor (FZD7) is enriched in LGR5+ intestinal stem cells and plays a critical role in their self-renewal. Yet, drug discovery approaches and structural bases for targeting specific FZD isoforms remain poorly defined. FZD proteins interact with Wnt signaling proteins via, in part, a lipid-binding groove on the extracellular cysteine-rich domain (CRD) of the FZD receptor. Here we report the identification of a potent peptide that selectively binds to the FZD7 CRD at a previously uncharacterized site and alters the conformation of the CRD and the architecture of its lipid-binding groove. Treatment with the FZD7-binding peptide impaired Wnt signaling in cultured cells and stem cell function in intestinal organoids. Together, our data illustrate that targeting the lipid-binding groove holds promise as an approach for achieving isoform-selective FZD receptor inhibition.


Asunto(s)
Receptores Frizzled/antagonistas & inhibidores , Receptores Frizzled/metabolismo , Intestinos/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Sitios de Unión , Células CHO , Membrana Celular/metabolismo , Cricetulus , Cristalografía por Rayos X , Descubrimiento de Drogas , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Intestinos/citología , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Péptidos/química , Unión Proteica , Multimerización de Proteína , Regeneración , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Células Madre/patología , Resonancia por Plasmón de Superficie , Vía de Señalización Wnt
5.
Nat Chem Biol ; 14(9): 902, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29728602

RESUMEN

The version of this article originally published contained older versions of the Life Sciences Reporting Summary and the Supplementary Text and Figures. The error has been corrected in the HTML and PDF versions of the article.

6.
Sci Signal ; 17(819): eabn1101, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227684

RESUMEN

Ubiquitination is a posttranslational modification that is crucial for the dynamic regulation of diverse signaling pathways. To enhance our understanding of ubiquitination-mediated signaling, we generated a new class of bispecific antibodies that combine recognition of ubiquitination substrates and specific polyubiquitin linkages. RIP1-K63 and RIP1-linear (Lin) linkage polyubiquitin bispecific antibodies detected linkage-specific ubiquitination of the proinflammatory kinase RIP1 in cells and in tissues and revealed RIP1 ubiquitination by immunofluorescence. Similarly, ubiquitination of the RIP1-related kinase RIP2 with K63 or linear linkages was specifically detected with the RIP2-K63 and RIP2-Lin bispecific antibodies, respectively. Furthermore, using the RIP2-K63 and RIP2-Lin bispecific antibodies, we found prominent K63-linked and linear RIP2 ubiquitination in samples from patients with ulcerative colitis and Crohn's disease. We also developed a bispecific antibody (K63-Lin) that simultaneously recognizes K63-linked and linear ubiquitination of components of various signaling pathways. Together, these bispecific antibodies represent a new class of reagents with the potential to be developed for the detection of inflammatory biomarkers.


Asunto(s)
Anticuerpos Biespecíficos , Ubiquitina , Humanos , Anticuerpos Biespecíficos/metabolismo , Poliubiquitina/metabolismo , Transducción de Señal/fisiología , Ubiquitina/metabolismo , Ubiquitinación
7.
Cell Death Differ ; 28(3): 985-1000, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32999468

RESUMEN

Receptor-interacting protein 1 (RIP1; RIPK1) is a key regulator of multiple signaling pathways that mediate inflammatory responses and cell death. TNF-TNFR1 triggered signaling complex formation, subsequent NF-κB and MAPK activation and induction of cell death involve RIPK1 ubiquitination at several lysine residues including Lys376 and Lys115. Here we show that mutating the ubiquitination site K376 of RIPK1 (K376R) in mice activates cell death resulting in embryonic lethality. In contrast to Ripk1K376R/K376R mice, Ripk1K115R/K115R mice reached adulthood and showed slightly higher responsiveness to TNF-induced death. Cell death observed in Ripk1K376R/K376R embryos relied on RIPK1 kinase activity as administration of RIPK1 inhibitor GNE684 to pregnant heterozygous mice effectively blocked cell death and prolonged survival. Embryonic lethality of Ripk1K376R/K376R mice was prevented by the loss of TNFR1, or by simultaneous deletion of caspase-8 and RIPK3. Interestingly, elimination of the wild-type allele from adult Ripk1K376R/cko mice was tolerated. However, adult Ripk1K376R/cko mice were exquisitely sensitive to TNF-induced hypothermia and associated lethality. Absence of the K376 ubiquitination site diminished K11-linked, K63-linked, and linear ubiquitination of RIPK1, and promoted the assembly of death-inducing cellular complexes, suggesting that multiple ubiquitin linkages contribute to the stability of the RIPK1 signaling complex that stimulates NF-κB and MAPK activation. In contrast, mutating K115 did not affect RIPK1 ubiquitination or TNF stimulated NF-κB and MAPK signaling. Overall, our data indicate that selective impairment of RIPK1 ubiquitination can lower the threshold for RIPK1 activation by TNF resulting in cell death and embryonic lethality.


Asunto(s)
Muerte Celular/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Ubiquitinación/efectos de los fármacos , Animales , Caspasa 8/metabolismo , Muerte Celular/genética , Desarrollo Embrionario/genética , Femenino , Quinasa I-kappa B/metabolismo , Inflamación/genética , Inflamación/metabolismo , Isoquinolinas/farmacología , Ratones , FN-kappa B/metabolismo , Fosforilación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Ubiquitinación/genética
8.
Elife ; 82019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31453810

RESUMEN

Upon detecting endoplasmic reticulum (ER) stress, the unfolded protein response (UPR) orchestrates adaptive cellular changes to reestablish homeostasis. If stress resolution fails, the UPR commits the cell to apoptotic death. Here we show that in hematopoietic cells, including multiple myeloma (MM), lymphoma, and leukemia cell lines, ER stress leads to caspase-mediated cleavage of the key UPR sensor IRE1 within its cytoplasmic linker region, generating a stable IRE1 fragment comprising the ER-lumenal domain and transmembrane segment (LDTM). This cleavage uncouples the stress-sensing and signaling domains of IRE1, attenuating its activation upon ER perturbation. Surprisingly, LDTM exerts negative feedback over apoptotic signaling by inhibiting recruitment of the key proapoptotic protein BAX to mitochondria. Furthermore, ectopic LDTM expression enhances xenograft growth of MM tumors in mice. These results uncover an unexpected mechanism of cross-regulation between the apoptotic caspase machinery and the UPR, which has biologically significant consequences for cell survival under ER stress.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Humanos , Ratones , Proteolisis
9.
Arterioscler Thromb Vasc Biol ; 24(4): 787-92, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15061151

RESUMEN

OBJECTIVE: The platelet-derived growth factor (PDGF) family consists of four members, PDGF A, PDGF B, and 2 new members, PDGF C and PDGF D, which signal through the alpha and beta PDGF receptor (PDGFR) tyrosine kinases. This study was performed to determine the receptor specificity and cellular expression profile of PDGF C. METHODS AND RESULTS: PDGF C growth factor domain (GFD) was shown to preferentially bind and activate alpha PDGFR and activate beta PDGFR when it is co-expressed with alpha PDGFR through heterodimer formation. An investigation of PDGF C mRNA and protein expression revealed that during mouse fetal development, PDGF C was expressed in the mesonephric mesenchyme, prefusion skeletal muscle, cardiac myoblasts, and in visceral and vascular smooth muscle, whereas in adult human tissues expression was largely restricted to smooth muscle. Microarray analysis of various cell types showed PDGF C expression in vascular smooth muscle cells, renal mesangial cells, and platelets. PDGF C mRNA expression in platelets was confirmed by real-time polymerase chain reaction, and PDGF C protein was localized in alpha granules by immuno-gold electron microscopy. Western blot analysis of platelets identified 55-kDa and 80-kDa PDGF C isoforms that were secreted on platelet activation. CONCLUSIONS: Taken together, our results demonstrated for the first time to our knowledge that like PDGF A and B, PDGF C is likely to play a role in platelet biology.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Plaquetas/metabolismo , Plaquetas/ultraestructura , Línea Celular/metabolismo , Gránulos Citoplasmáticos/química , ADN Complementario/genética , Dimerización , Desarrollo Embrionario y Fetal , Endopeptidasas/sangre , Humanos , Linfocinas , Ratones/embriología , Ratones Endogámicos BALB C , Músculo Liso Vascular/metabolismo , Especificidad de Órganos , Fosforilación , Activación Plaquetaria , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-sis/química , Proteínas Proto-Oncogénicas c-sis/metabolismo , ARN Mensajero/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Transfección
10.
Methods Mol Biol ; 289: 157-70, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15502181

RESUMEN

HOX homeodomain proteins are thought to be master developmental regulators of tissue patterning during embryogenesis. These DNA binding proteins also have diverse roles in adult cell function, and derangement of HOX genes has been associated with several types of cancer. In this chapter we present protocols for the immunohistochemical localization of HOX proteins in the epidermis. We also provide in situ hybridization protocols for detection of HOX gene mRNA transcripts in the epidermis.


Asunto(s)
Células Epidérmicas , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , ARN Mensajero/genética , Animales , Epidermis/metabolismo , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Queratinocitos
11.
Biochem J ; 379(Pt 2): 385-93, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-14713286

RESUMEN

We have identified a novel membrane form of guanylate cyclase (GC) from a mouse testis cDNA library and termed it mGC-G (mouse GC-G) based on its high sequence homology to rat GC-G. It encodes a potential type I transmembrane receptor, with the characteristic domain structure common to all members of the family of membrane GCs, including an extracellular, putative ligand-binding domain, a single membrane-spanning segment and cytoplasmic protein kinase-like and cyclase catalytic domains. Real-time quantitative reverse transcriptase--PCR and Northern-blot analyses showed that mGC-G is highly and selectively expressed in mouse testis. Phylogenetic analysis based on the extracellular protein sequence revealed that mGC-G is closely related to members of the subfamily of natriuretic peptide receptor GCs. When overexpressed in HEK-293T cells (human embryonic kidney 293T cells) or COS-7 cells, mGC-G manifests as a membrane-bound glycoprotein, which can form either homomeric or heteromeric complexes with the natriuretic peptide receptor GC-A. It exhibits marked cGMP-generating GC activity; however, notably, all ligands known to activate other receptor GCs failed to stimulate enzymic activity. The unique testis-enriched expression of mGC-G, which is completely different from the broader tissue distribution of rat GC-G, suggests the existence of as-yet-unidentified ligands and unappreciated species-specific physiological functions mediated through mGC-G/cGMP signalling in the testis.


Asunto(s)
Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Testículo/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Clonación Molecular , GMP Cíclico/metabolismo , Expresión Génica , Glicosilación , Guanilato Ciclasa/clasificación , Humanos , Masculino , Proteínas de la Membrana/clasificación , Ratones , Datos de Secuencia Molecular , Péptidos Natriuréticos/farmacología , Filogenia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Distribución Tisular
12.
Physiol Genomics ; 11(2): 99-107, 2002 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-12388791

RESUMEN

We have developed a system utilizing the murine Tie2 promoter/enhancer coupled with the "tetracycline-on" regulatory elements to create a model that allows regulated and selective expression of a beta-galactosidase (betaGal) reporter transgene in the adult murine vascular endothelium. Two independent lines of viable and fertile mice were characterized, and they exhibit minimal betaGal expression under basal conditions. In response to exogenous doxycycline (Dox), selective expression of betaGal was demonstrated in the vascular endothelium of all tissues examined. En face analyses of the aorta and its principle branches indicate that the vast majority of lumenal endothelial cells express the transgene. Inducible betaGal expression also extends to the endocardium and the microvasculature of all organs. There is no evidence of specific transgene expression in nonendothelial cell types. Induction of the betaGal was effectively achieved after 3 days of oral Dox treatment and persisted for over 3 mo with continuous administration. This model can now be widely applied to study the role of specific genes in the phenotype of adult murine vasculature.


Asunto(s)
Endotelio Vascular/química , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Transgenes/genética , Animales , Elementos de Facilitación Genéticos/genética , Vectores Genéticos/genética , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Factores R/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2 , Proteínas Represoras/genética , beta-Galactosidasa/genética
13.
Physiol Genomics ; 12(1): 13-23, 2002 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-12419857

RESUMEN

Biomechanical forces can modulate endothelial phenotype through changes in gene expression. We hypothesized that physiological laminar shear stresses (LSS) act as differentiative stimuli on endothelial cells (EC) to alter gene expression, creating an antioxidant, anti-apoptotic and anti-proliferative environment. The transcriptional profile of cultured human umbilical vein endothelial cells (HUVEC) exposed to LSS was evaluated by GeneCalling; 107 genes demonstrated at least a twofold change in expression at 24 h (LSS vs. static). These flow-responsive genes represent a limited number of functional clusters that include transcription factors, antioxidants, signaling molecules, cell cycle regulators, and genes involved in cellular differentiation. Immunohistochemistry and in situ hybridization confirmed that many of these flow-responsive genes, including the novel basic helix-loop-helix transcription factor Hath6, are expressed in EC in vivo. Thus these data identify a limited set of flow-responsive genes expressed in the endothelium that may be responsible for the establishment and maintenance of the flow-adapted endothelial phenotype in vivo.


Asunto(s)
Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica/métodos , Transcripción Genética , Secuencia de Aminoácidos , Antioxidantes/metabolismo , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Ciclo Celular , Diferenciación Celular , División Celular , Células Cultivadas , Citocinas/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Estrés Mecánico , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Regulación hacia Arriba
14.
J Invest Dermatol ; 118(1): 25-34, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11851872

RESUMEN

Liver X receptor-alpha and -beta are members of the nuclear hormone receptor superfamily that heterodimerize with retinoid X receptor and are activated by oxysterols. In recent studies we found that treatment of cultured human keratinocytes with oxysterolstimulated differentiation, as demonstrated by increased expression of involucrin and transglutaminase, and inhibited proliferation. The aims of this study were to determine: (i) whether oxysterols applied topically to the skin of mice induce differentiation in normal epidermis; (ii) whether this effect is mediated via liver X receptor-alpha and/or liver X receptor-beta; and (iii) whether oxysterols normalize epidermal morphology in an animal model of epidermal hyperplasia. Topical treatment of normal hairless mice with 22(R)-hydroxycholesterol or 24(S),25-epoxycholesterol resulted in a decrease in epidermal thickness and a decrease in keratinocyte proliferation assayed by proliferating cell nuclear antigen staining. Moreover, oxysterol treatment increased the levels of involucrin, loricrin, and profilaggrin protein and mRNA in the epidermis, indicating that oxysterols stimulate epidermal differentiation. Additionally, topical oxysterol pretreatment improved permeability barrier homeostasis. Whereas liver X receptor-alpha-/- mice revealed no alterations in epidermal differentiation, the epidermis was thinner in liver X receptor-beta-/- mice than in wild-type mice, with a reduced number of proliferating cell nuclear antigen positive cells and a modest reduction in the expression of differentiation markers. Topical oxysterol treatment induced differentiation in liver X receptor-alpha-/- mice whereas in liver X receptor-beta-/- mice there was no increase in the expression of differentiation markers. Whereas both liver X receptor-alpha and liver X receptor-beta are expressed in cultured human keratinocytes and in fetal rat skin, only liver X receptor-beta was observed on northern blotting in adult mouse epidermis. Finally, treatment of hyperproliferative epidermis with oxysterols restored epidermal homeostasis. These studies demonstrate that epidermal differentiation is regulated by liver X receptor-beta and that oxysterols, acting via liver X receptor-beta, can induce differentiation and inhibit proliferation in vivo. The ability of oxysterols to reverse epidermal hyperplasia suggests that these agents could be beneficial for the treatment of skin disorders associated with hyperproliferation and/or altered differentiation.


Asunto(s)
Colesterol/farmacología , Células Epidérmicas , Epidermis/efectos de los fármacos , Hidroxicolesteroles/farmacología , Hígado/metabolismo , Receptores de Ácido Retinoico/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Colesterol/análogos & derivados , Epidermis/metabolismo , Epidermis/patología , Homeostasis/efectos de los fármacos , Hiperplasia , Queratinocitos/citología , Ratones , Ratones Pelados , Permeabilidad/efectos de los fármacos , Isoformas de Proteínas/fisiología , Receptores de Ácido Retinoico/deficiencia , Receptores X Retinoide , Factores de Transcripción/deficiencia
15.
Sci Signal ; 5(216): ra22, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22434933

RESUMEN

Tumor necrosis factor (TNF) family members are essential for the development and proper functioning of the immune system. TNF receptor (TNFR) signaling is mediated through the assembly of protein signaling complexes that activate the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in a ubiquitin-dependent manner. The cellular inhibitor of apoptosis (c-IAP) proteins c-IAP1 and c-IAP2 are E3 ubiquitin ligases that are recruited to TNFR signaling complexes through their constitutive association with the adaptor protein TNFR-associated factor 2 (TRAF2). We demonstrated that c-IAP1 and c-IAP2 were required for canonical activation of NF-κB and MAPK by members of the TNFR family. c-IAPs were required for the recruitment of inhibitor of κB kinase ß (IKKß), the IKK regulatory subunit NF-κB essential modulator (NEMO), and RBCK1/Hoil1-interacting protein (HOIP) to TNFR signaling complexes and the induction of gene expression by TNF family members. In contrast, TNFRs that stimulated the noncanonical NF-κB pathway triggered translocation of c-IAPs, TRAF2, and TRAF3 from the cytosol to membrane fractions, which led to their proteasomal and lysosomal degradation. Finally, we established that signaling by B cell-activating factor receptor 3 induced the cytosolic depletion of TRAF3, which enabled noncanonical NF-κB activation. These results define c-IAP proteins as critical regulators of the activation of NF-κB and MAPK signaling pathways by members of the TNFR superfamily.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/inmunología , Factores de Necrosis Tumoral/metabolismo , Western Blotting , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Silenciador del Gen , Humanos , Quinasa I-kappa B/metabolismo , Proteínas Inhibidoras de la Apoptosis/inmunología , Transporte de Proteínas , ARN Interferente Pequeño/genética , Receptores de Interleucina-4/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factores de Necrosis Tumoral/inmunología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas
16.
Nat Cell Biol ; 14(1): 61-72, 2011 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-22179047

RESUMEN

Using RNAi screening, proteomics, cell biological and mouse genetics approaches, we have identified a complex of nine proteins, seven of which are disrupted in human ciliopathies. A transmembrane component, TMEM231, localizes to the basal body before and independently of intraflagellar transport in a Septin 2 (Sept2)-regulated fashion. The localizations of TMEM231, B9D1 (B9 domain-containing protein 1) and CC2D2A (coiled-coil and C2 domain-containing protein 2A) at the transition zone are dependent on one another and on Sept2. Disruption of the complex in vitro causes a reduction in cilia formation and a loss of signalling receptors from the remaining cilia. Mouse knockouts of B9D1 and TMEM231 have identical defects in Sonic hedgehog (Shh) signalling and ciliogenesis. Strikingly, disruption of the complex increases the rate of diffusion into the ciliary membrane and the amount of plasma-membrane protein in the cilia. The complex that we have described is essential for normal cilia function and acts as a diffusion barrier to maintain the cilia membrane as a compartmentalized signalling organelle.


Asunto(s)
Cilios/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , Proteínas del Citoesqueleto , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Proteínas/metabolismo , Septinas/metabolismo , Transducción de Señal
17.
Proc Natl Acad Sci U S A ; 102(34): 12077-82, 2005 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-16105945

RESUMEN

Enzymes of the blood coagulation pathway enhance the inflammatory response leading to endothelial dysfunction, accounting, in part, for the vascular complications occurring in sepsis and cardiovascular disease. The responses of endothelial cell activation include induction of the expression of tissue factor (TF), a membrane glycoprotein that promotes thrombosis, and of E-selectin, a cell adhesion molecule that promotes inflammation. In this report, we demonstrate synergistic interactions between the coagulation factor Xa (fXa) and the proinflammatory cytokines TNF, IL-1beta, and CD40L, leading to enhanced expression of TF and E-selectin in endothelial cells. A detailed analysis of the molecular pathways that could account for this activity of fXa showed that fXa inhibited the cytokine-induced expression of dual specificity phosphatases, MAP kinase phosphatase-L, -4, -5, and -7, blocking a negative regulatory effect on c-Jun N-terminal kinase. The synergistic interaction between fXa and TNF was also involved in the inhibition of A20 and IkappaBalpha expression in the IkappaB kinase-NF-kappaB pathway. The data indicate that inhibition of negative regulatory signaling accounts for the amplification of cytokine-induced endothelial cell activation by fXa.


Asunto(s)
Coagulación Sanguínea/fisiología , Células Endoteliales/metabolismo , Factor Xa/metabolismo , Regulación de la Expresión Génica , Transducción de Señal/fisiología , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ligando de CD40/metabolismo , Cartilla de ADN , Selectina E/metabolismo , Células Endoteliales/fisiología , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Interleucina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Dev Dyn ; 227(2): 192-202, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12761847

RESUMEN

Substantial evidence suggests that HOX homeobox genes regulate aspects of body development, including hair formation. We initially isolated the HOXB13 gene from human fetal skin in experiments designed to identify candidate genes that regulate scarless fetal wound healing. Although the HOX homeodomain proteins have been proposed to function as transcription factors, we have demonstrated previously that substantial fractions of the HOXB6 and HOXB4 proteins are localized to the cytoplasm throughout epidermal development. The purpose of the current study was to identify HOXB13 protein expression patterns in developing skin to elucidate potential mechanisms by which this protein might regulate aspects of tissue development and healing. HOXB13 protein expression was detected throughout the developing epidermis, with weaker signal observed in the early developing dermis. Epidermal HOXB13 signal was detected over the entire body surface, but surprisingly, essentially all of the signal was cytoplasmic in developing skin. Low-level HOXB13 protein expression was detected in adult skin and within the telogen hair follicle, and a portion of the residual signal in adult epidermis was nuclear. Expression in hyperproliferative skin conditions remained cytoplasmic with the exception of epidermis associated with Kaposi's sarcoma, which showed strong HOXB13 expression that was partially localized to the nucleus.


Asunto(s)
Epidermis/embriología , Epidermis/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Animales , Cromosomas , Citoplasma/metabolismo , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/embriología , Folículo Piloso/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedades de la Piel/fisiopatología
19.
Dev Dyn ; 224(1): 58-68, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11984874

RESUMEN

The HOX homeodomain proteins are fundamental regulators of organ and tissue development, where they are thought to function as transcription factors, and HOX gene expression has been associated with numerous types of cancers. Previous studies have demonstrated that enforced expression of the HOXB4 protein transforms cultured fibroblasts and leads to a selective expansion of the hematopoietic stem cell pool, suggesting that this protein might play a role in cellular proliferation. In support of this concept, we now show that enforced expression of HOXB4 in human neonatal keratinocytes results in increased cellular proliferation and colony formation as well as decreased expression of the alpha-2-integrin and CD44 cell surface adhesion molecules. We previously have reported HOXB4 gene expression in the basal and suprabasal layers of developing human skin and now show extensive HOXB4 mRNA in psoriatic skin and basal cell carcinoma. In fetal human skin HOXB4 protein expression was both nuclear and cytoplasmic within epidermal basal cells and in hair follicle inner and outer root sheath cells, whereas strong nuclear signals were observed in the bulge region. In adult skin, HOXB4 protein expression was both nuclear and cytoplasmic, but was predominantly localized to the intermediate and differentiated cell layers. In contrast to the striking gradient patterns of HOX gene and protein expression previously described in developing spinal cord and limb, HOXB4 protein was uniformly detected in all regions of the fetal and adult skin. Although little HOXB4 signal localized to proliferative cell layers, as marked by proliferating cell nuclear antigen (PCNA) staining, in normal adult epidermis, nuclear HOXB4 protein expression substantially overlapped with PCNA-positive cell in a series of samples of hyperproliferative skin. Taken together, these data suggest that nuclear HOXB4 protein may play a role in the regulation of cellular proliferation/adhesion in developing fetal human epidermis and in hyperproliferation conditions, including cancers, in adult epidermis. Published 2002 Wiley-Liss, Inc.


Asunto(s)
Epidermis/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Queratinocitos/fisiología , Enfermedades de la Piel/metabolismo , Factores de Transcripción/metabolismo , Adulto , Antígenos CD/metabolismo , Adhesión Celular , División Celular , Núcleo Celular/metabolismo , Células Epidérmicas , Epidermis/embriología , Epidermis/metabolismo , Genes Homeobox , Proteínas de Homeodominio/genética , Humanos , Receptores de Hialuranos/metabolismo , Hibridación in Situ , Integrina alfa2 , Queratinocitos/citología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Transcripción/genética
20.
J Biol Chem ; 278(35): 33232-8, 2003 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12807873

RESUMEN

We have previously utilized a combination of high throughput sequencing and genome-wide microarray profiling analyses to identify novel cell-surface proteins expressed in human umbilical vein endothelial cells. One gene identified by this approach encodes a type I transmembrane receptor that shares sequence homology with the intracellular domain of members of the interleukin-17 (IL-17) receptor family. Real-time quantitative PCR and Northern analyses revealed that this gene is highly expressed in human umbilical vein endothelial cells and in several highly vascularized tissues such as kidney, colon, skeletal muscle, heart, and small intestine. In addition, we also found that it is also highly expressed in the ductal epithelial cells of human salivary glands, seminal vesicles, and the collecting tubules of the kidney by in situ hybridization. This putative receptor, which we have termed human SEF (hSEF), is also expressed in a variety of breast cancer tissues. In co-immunoprecipitation assays, this receptor is capable of forming homomeric complexes and can interact with fibroblast growth factor (FGF) receptor 1. Overexpression of this receptor inhibits FGF induction of an FGF-responsive reporter gene in human 293T cells. This appears to occur as a result of specific inhibition of p42/p44 ERK in the absence of upstream MEK inhibition. This inhibitory effect is dependent upon a functional intracellular domain since deletion mutants missing the IL-17 receptor-like domain lack this inhibitory effect. These findings are consistent with the recent discovery of the zebrafish homologue, Sef (similar expression to fgf genes), which specifically antagonizes FGF signaling when ectopically expressed in zebrafish or Xenopus laevis embryos. Based on sequence and functional similarities, this novel IL-17 receptor homologue represents a potential human SEF and is likely to play critical roles in endothelial or epithelial functions such as proliferation, migration, and angiogenesis.


Asunto(s)
Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Proteínas Recombinantes/química , Transducción de Señal , Venas Umbilicales/citología , Secuencia de Aminoácidos , Animales , Northern Blotting , Western Blotting , División Celular , Línea Celular , Movimiento Celular , Células Cultivadas , Clonación Molecular , Genes Reporteros , Humanos , Hibridación in Situ , Luciferasas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Neovascularización Fisiológica , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina-17 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Distribución Tisular , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA