Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34810256

RESUMEN

Global inactivation of IκB kinase (IKK)-α results in defective lymph node (LN) formation and B cell maturation, and loss of IKK-α-dependent noncanonical NF-κB signaling in stromal organizer and hematopoietic cells is thought to underlie these distinct defects. We previously demonstrated that this pathway is also activated in vascular endothelial cells (ECs). To determine the physiologic function of EC-intrinsic IKK-α, we crossed IkkαF/F mice with Tie2-cre or Cdh5-cre mice to ablate IKK-α in ECs. Notably, the compound defects of global IKK-α inactivation were recapitulated in IkkαTie2 and IkkαCdh5 mice, as both lacked all LNs and mature follicular and marginal zone B cell numbers were markedly reduced. However, as Tie2-cre and Cdh5-cre are expressed in all ECs, including blood forming hemogenic ECs, IKK-α was also absent in hematopoietic cells (HC). To determine if loss of HC-intrinsic IKK-α affected LN development, we generated IkkαVav mice lacking IKK-α in only the hematopoietic compartment. While mature B cell numbers were significantly reduced in IkkαVav mice, LN formation was intact. As lymphatic vessels also arise during development from blood ECs, we generated IkkαLyve1 mice lacking IKK-α in lymphatic ECs (LECs) to determine if IKK-α in lymphatic vessels impacts LN development. Strikingly, while mature B cell numbers were normal, LNs were completely absent in IkkαLyve1 mice. Thus, our findings reveal that IKK-α in distinct EC-derived compartments is uniquely required to promote B cell homeostasis and LN development, and we establish that LEC-intrinsic IKK-α is absolutely essential for LN formation.


Asunto(s)
Linfocitos B/metabolismo , Quinasa I-kappa B/fisiología , Ganglios Linfáticos/metabolismo , Animales , Linfocitos B/fisiología , Línea Celular , Células Endoteliales/metabolismo , Femenino , Homeostasis/fisiología , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Ganglios Linfáticos/fisiología , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Organogénesis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Biol Chem ; 291(16): 8440-52, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26826124

RESUMEN

T cell activation following antigen binding to the T cell receptor (TCR) involves the mobilization of intracellular Ca(2+) to activate the key transcription factors nuclear factor of activated T lymphocytes (NFAT) and NF-κB. The mechanism of NFAT activation by Ca(2+) has been determined. However, the role of Ca(2+) in controlling NF-κB signaling is poorly understood, and the source of Ca(2+) required for NF-κB activation is unknown. We demonstrate that TCR- but not TNF-induced NF-κB signaling upstream of IκB kinase activation absolutely requires the influx of extracellular Ca(2+) via STIM1-dependent Ca(2+) release-activated Ca(2+)/Orai channels. We further show that Ca(2+) influx controls phosphorylation of the NF-κB protein p65 on Ser-536 and that this posttranslational modification controls its nuclear localization and transcriptional activation. Notably, our data reveal that this role for Ca(2+) is entirely separate from its upstream control of IκBα degradation, thereby identifying a novel Ca(2+)-dependent distal step in TCR-induced NF-κB activation. Finally, we demonstrate that this control of distal signaling occurs via Ca(2+)-dependent PKCα-mediated phosphorylation of p65. Thus, we establish the source of Ca(2+) required for TCR-induced NF-κB activation and define a new distal Ca(2+)-dependent checkpoint in TCR-induced NF-κB signaling that has broad implications for the control of immune cell development and T cell functional specificity.


Asunto(s)
Canales de Calcio/biosíntesis , Señalización del Calcio/fisiología , Calcio/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional/fisiología , Canales de Calcio/genética , Humanos , Células Jurkat , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Proteína ORAI1 , Fosforilación/fisiología , Receptores de Antígenos de Linfocitos T/genética , Molécula de Interacción Estromal 1 , Factor de Transcripción ReIA/genética
3.
Biochem Biophys Res Commun ; 450(1): 341-6, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24942881

RESUMEN

Non-canonical NF-κB signaling is controlled by the precise regulation of NF-κB inducing kinase (NIK) stability. NIK is constitutively ubiquitylated by cellular inhibitor of apoptosis (cIAP) proteins 1 and 2, leading to its complete proteasomal degradation in resting cells. Following stimulation, cIAP-mediated ubiquitylation of NIK ceases and NIK is stabilized, allowing for inhibitor of κB kinase (IKK)α activation and non-canonical NF-κB signaling. Non-canonical NF-κB signaling is terminated by feedback phosphorylation of NIK by IKKα that promotes NIK degradation; however, the mechanism of active NIK protein turnover remains unknown. To address this question, we established a strategy to precisely distinguish between basal degradation of newly synthesized endogenous NIK and induced active NIK in stimulated cells. Using this approach, we found that IKKα-mediated degradation of signal-induced activated NIK occurs through the proteasome. To determine whether cIAP1 or cIAP2 play a role in active NIK turnover, we utilized a Smac mimetic (GT13072), which promotes degradation of these E3 ubiquitin ligases. As expected, GT13072 stabilized NIK in resting cells. However, loss of the cIAPs did not inhibit proteasome-dependent turnover of signal-induced NIK showing that unlike the basal regulatory mechanism, active NIK turnover is independent of cIAP1 and cIAP2. Our results therefore establish that the negative feedback control of IKKα-mediated NIK turnover occurs via a novel proteasome-dependent and cIAP-independent mechanism.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Regulación de la Expresión Génica/fisiología , Quinasa I-kappa B/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células 3T3 , Animales , Activación Enzimática , Células HeLa , Humanos , Ratones , Quinasa de Factor Nuclear kappa B
4.
Transplant Cell Ther ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39084263

RESUMEN

Allogeneic hematopoietic stem cell transplant (HSCT) remains the only potentially curative treatment for many hematologic malignancies (HM). We previously developed a two-step approach that separates the lymphoid and myeloid portions of the graft, allowing a consistent T cell dosing and sparing the stem cells from the effect of post-transplant cyclophosphamide (CY). The two-step approach demonstrated safety and efficacy in patients treated with myeloablative and reduced-intensity conditioning. Here, we extended our two-step platform to older and less fit patients and explored the effects of using a high dose of T cells on disease relapse and transplant outcomes. Thirty-four patients with HM were treated. Median age was 68 years old and included a minority population constituting 32%. Eighty-two percent had a hematopoietic cell transplantation comorbidity index score ≥3. Ninety-one percent were haploidentical, and the rest were matched-related donor HSCT. Following administration of fludarabine and 2 Gy total body irradiation (TBI) (13 patients) or 4 Gy TBI (21 patients) conditioning regimen, a fixed dose of 2 × 108/kg CD3+ T cells was given, followed 2 days later by CY, then infusion of CD34-selected stem cells. Overall survival (OS) was 70% at 1 year and 48% at 3 years. The cumulative incidence (CI) of non-relapse mortality (NRM) and relapse were 22% and 33% at 3 years. However, the CI of relapse was much lower for patients treated with 4 Gy TBI versus those treated with 2 Gy TBI (11% versus 54%, P = .045), while NRM was similar (23% versus 15%, P = .399). This contributed to a high OS of 64% in patients who received 4 Gy TBI-based conditioning at 3 years, with median OS not reached, although this was not statistically significant (P = .68). The median time to neutrophil and platelet recovery was 12 and 17 days, respectively. The CI of grade II acute graft-versus-host-disease (aGVHD) was 22% and 26% at 100 days and 6 months, respectively. The CI of chronic GVHD (cGVHD) was 7.5% at 3 years. There was no grade III or IV aGVHD, no severe cGVHD, and no deaths attributable to GVHD. In conclusion, the two-step approach HSCT demonstrated a low disease relapse rate and high survival in patients treated with 4 Gy TBI-based conditioning, despite a generally older and more medically compromised patient population.

5.
J Immunol ; 185(5): 2665-9, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20693425

RESUMEN

The signaling and adaptor protein Homer3 plays a role in controlling immune homeostasis and self-reactivity. Homer3 is recruited to the immune synapse (IS) following TCR ligation, although the mechanisms regulating this subcellular localization are unknown. We show that Homer3 specifically associates with a novel ubiquitin-like domain in the IkappaB kinase (IKK) beta subunit of the IKK complex. Homer3 associates with IKKbeta in T cells and colocalizes with the IKK complex at the IS. However, Homer3 is not required for IKK activation, as NF-kappaB signaling is intact in Homer3-deficient T cells. Instead, the IKK complex recruits Homer3 to the IS following TCR engagement, and we present evidence that this association regulates actin dynamics in T cells. These findings identify a novel interaction between two major signaling proteins and reveal an unexpected NF-kappaB-independent function for the IKK complex in regulating the subcellular localization of Homer3.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas I-kappa B/metabolismo , Proteínas Portadoras/fisiología , Línea Celular , Células HeLa , Proteínas de Andamiaje Homer , Humanos , Proteínas I-kappa B/fisiología , Sinapsis Inmunológicas/enzimología , Sinapsis Inmunológicas/inmunología , Células Jurkat , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología , Fracciones Subcelulares/inmunología , Fracciones Subcelulares/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo
6.
Oncogene ; 24(9): 1625-33, 2005 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-15674328

RESUMEN

This study aimed to (a) determine if DNA methylation is a mechanism of WWOX (WW domain containing oxidoreductase) and FHIT (fragile histidine triad) inactivation in lung, breast and bladder cancers; (b) examine distinct methylation patterns in neoplastic and adjacent tissues and (c) seek correlation of methylation patterns with disease status. Protein expression was detected by immunohistochemistry, and methylation status by methylation-specific PCR (MSP) and sequencing, in lung squamous cell carcinomas and adjacent tissues, invasive breast carcinomas, adjacent tissues and normal mammary tissues and bladder transitional cell carcinomas. Wwox and Fhit expression was reduced in cancers in association with hypermethylation. Differential patterns of WWOX and FHIT methylation were observed in neoplastic vs adjacent non-neoplastic tissues, suggesting that targeted MSP amplification could be useful in following treatment or prevention protocols. WWOX promoter MSP differentiates DNA of lung cancer from DNA of adjacent lung tissue. WWOX and FHIT promoter methylation is detected in tissue adjacent to breast cancer and WWOX exon 1 MSP distinguishes breast cancer DNA from DNA of adjacent and normal tissue. Differential methylation in cancerous vs adjacent tissues suggests that WWOX and FHIT hypermethylation analyses could enrich a panel of DNA methylation markers.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Neoplasias de la Mama/genética , Fragilidad Cromosómica/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Oxidorreductasas/genética , Neoplasias de la Vejiga Urinaria/genética , Apoptosis/genética , Secuencia de Bases , Neoplasias de la Mama/patología , Metilación de ADN , Cartilla de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor , Neoplasias de la Vejiga Urinaria/patología , Oxidorreductasa que Contiene Dominios WW
7.
Cancer Lett ; 232(1): 27-36, 2006 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-16225988

RESUMEN

It was hypothesized as early as 1986, that the recently discovered common fragile sites could facilitate recombination events, such as deletions and translocations, that result in clonally expanded cancer cell populations with specific chromosome alterations in specific cancer types. A natural extension of this hypothesis is that the clonal expansion must be driven by alteration of genes at recombination breakpoints whose altered functions actually drive clonal expansion. Nevertheless, when the FHIT gene was discovered at FRA3B, the most active common chromosome fragile region, and proposed as an example of a tumor suppressor gene altered by chromosome translocations and deletions, a wave of reports suggested that the FHIT gene was altered in cancer simply because it was in a fragile region and not because it had contributed to the clonal expansion, thus turning the original hypothesis upside down. Now, after nearly ten years and more than 500 FHIT reports, it is apparent that FHIT is an important tumor suppressor gene and that there are genes at other fragile regions that contribute significantly to development of cancer. A second fragile gene with a demonstrated role in cancer development is the WWOX gene on chromosome 16q; alterations to the WWOX gene contribute to development of hormone responsive and other cancers. Results of our recent studies of these two fragile tumor suppressor genes were summarized at the first Fragilome meeting in Heidelberg, Feb. 2005.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Sitios Frágiles del Cromosoma/fisiología , Fragilidad Cromosómica , Proteínas de Neoplasias/genética , Neoplasias/genética , Oxidorreductasas/genética , Animales , Apoptosis , Cromosomas Humanos Par 16/genética , Epigénesis Genética , Humanos , Proteínas Supresoras de Tumor , Oxidorreductasa que Contiene Dominios WW
8.
Methods Mol Biol ; 1280: 155-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25736748

RESUMEN

NF-κB is a family of transcription factors regulated through two distinct signaling cascades, the classical and the Noncanonical NF-κB pathways. Noncanonical NF-κB plays important roles in the immune system, as it is necessary for lymphoid organogenesis and B-cell survival and differentiation, as well as osteoclastogenesis. In the last few years, there has been an increased number of studies focusing on both identifying the upstream events that regulate the noncanonical NF-κB pathway as well as determining the physiological roles of noncanonical NF-κB in normal and disease pathologies, such as cancer and autoimmune diseases. Dysregulation of noncanonical NF-κB has now been associated with the pathogenesis of several types of lymphomas and autoimmune diseases and is believed to contribute to chronic inflammatory diseases, including ulcerative colitis. These studies suggest that targeting the Noncanonical pathway, similar to classical NF-κB, may have some therapeutic potential in the future; however, there is still quite a bit about the regulation of the noncanonical signaling that remains to be defined. In this chapter we describe the use of HUVEC, as an in vitro model for examining noncanonical NF-κB signaling in response to different stimuli. We demonstrate two different methods to measure noncanonical NF-κB activation: the processing of p100 to p52, and noncanonical NF-κB-dependent gene expression of CXCL12. The first method examines a key regulatory requirement for noncanonical NF-κB activation, by which p100 undergoes proteolytic cleavage to relieve the inhibition of NF-κB dimers for nuclear translocation and activation of gene transcription. The latter demonstrates the downstream effects of activated noncanonical NF-κB in response to stimuli.


Asunto(s)
Quimiocina CXCL12/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , FN-kappa B/metabolismo , Activación Transcripcional , Linfocitos B/metabolismo , Western Blotting/métodos , Técnicas de Cultivo de Célula , Separación Celular/métodos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Subunidad p52 de NF-kappa B/metabolismo , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Methods Mol Biol ; 1280: 505-25, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25736769

RESUMEN

NF-κB comprises a family of transcription factors that regulate the expression of diverse gene families essential for inflammatory and immune responses as well as cell survival and cell death pathways. Aberrant NF-κB transcriptional activity plays pivotal roles in a large number of human pathologies, including a variety of cancers and chronic inflammatory diseases. Therefore, there has been a large increase in studies aimed at identifying and testing drugs or small molecule inhibitors that would specifically block NF-κB activation in inflammatory diseases and cancer. In this chapter, we describe an in vivo system to test the inhibitory effects of the NEMO-binding domain (NBD) peptide on NF-κB activation specifically in the vascular endothelium and lymphocytes in mice. We demonstrate that pretreatment of mice with the NBD peptide reduces the NF-κB induced gene expression of cell adhesion molecules and DNA-binding activity following systemic LPS stimulation. These methods can be further used to test alternate inhibitors for effects on NF-κB signaling in murine endothelium and immune cells.


Asunto(s)
Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/metabolismo , Fragmentos de Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos , Animales , Aorta/metabolismo , Western Blotting/métodos , Selectina E/metabolismo , Ensayo de Cambio de Movilidad Electroforética/métodos , Activación Enzimática/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/química , Lipopolisacáridos/administración & dosificación , Ratones , Fragmentos de Péptidos/administración & dosificación , Bazo/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
J Exp Med ; 212(10): 1513-28, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26371187

RESUMEN

Innate lymphoid cells (ILCs) are critical for maintaining epithelial barrier integrity at mucosal surfaces; however, the tissue-specific factors that regulate ILC responses remain poorly characterized. Using mice with intestinal epithelial cell (IEC)-specific deletions in either inhibitor of κB kinase (IKK)α or IKKß, two critical regulators of NFκB activation, we demonstrate that IEC-intrinsic IKKα expression selectively regulates group 3 ILC (ILC3)-dependent antibacterial immunity in the intestine. Although IKKß(ΔIEC) mice efficiently controlled Citrobacter rodentium infection, IKKα(ΔIEC) mice exhibited severe intestinal inflammation, increased bacterial dissemination to peripheral organs, and increased host mortality. Consistent with weakened innate immunity to C. rodentium, IKKα(ΔIEC) mice displayed impaired IL-22 production by RORγt(+) ILC3s, and therapeutic delivery of rIL-22 or transfer of sort-purified IL-22-competent ILCs from control mice could protect IKKα(ΔIEC) mice from C. rodentium-induced morbidity. Defective ILC3 responses in IKKα(ΔIEC) mice were associated with overproduction of thymic stromal lymphopoietin (TSLP) by IECs, which negatively regulated IL-22 production by ILC3s and impaired innate immunity to C. rodentium. IEC-intrinsic IKKα expression was similarly critical for regulation of intestinal inflammation after chemically induced intestinal damage and colitis. Collectively, these data identify a previously unrecognized role for epithelial cell-intrinsic IKKα expression and TSLP in regulating ILC3 responses required to maintain intestinal barrier immunity.


Asunto(s)
Quinasa I-kappa B/metabolismo , Inmunidad Innata/inmunología , Linfocitos/inmunología , Animales , Citrobacter rodentium/patogenicidad , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Citocinas/metabolismo , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/mortalidad , Células Epiteliales/metabolismo , Femenino , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Interleucinas/genética , Interleucinas/metabolismo , Interleucinas/farmacología , Linfocitos/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Linfopoyetina del Estroma Tímico , Interleucina-22
11.
Sci Signal ; 7(311): ra13, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24497610

RESUMEN

Precise regulation of nuclear factor κB (NF-κB) signaling is crucial for normal immune responses, and defective NF-κB activity underlies a range of immunodeficiencies. NF-κB is activated through two signaling cascades: the classical and noncanonical pathways. The classical pathway requires inhibitor of κB kinase ß (IKKß) and NF-κB essential modulator (NEMO), and hypomorphic mutations in the gene encoding NEMO (ikbkg) lead to inherited immunodeficiencies, collectively termed NEMO-ID. Noncanonical NF-κB activation requires NF-κB-inducing kinase (NIK) and IKKα, but not NEMO. We found that noncanonical NF-κB was basally active in peripheral blood mononuclear cells from NEMO-ID patients and that noncanonical NF-κB signaling was similarly enhanced in cell lines lacking functional NEMO. NIK, which normally undergoes constitutive degradation, was aberrantly present in resting NEMO-deficient cells, and regulation of its abundance was rescued by reconstitution with full-length NEMO, but not a mutant NEMO protein unable to physically associate with IKKα or IKKß. Binding of NEMO to IKKα was not required for ligand-dependent stabilization of NIK or noncanonical NF-κB signaling. Rather, an intact and functional IKK complex was essential to suppress basal NIK activity in unstimulated cells. Despite interacting with IKKα and IKKß to form an IKK complex, NEMO mutants associated with immunodeficiency failed to rescue classical NF-κB signaling or reverse the accumulation of NIK. Together, these findings identify a crucial role for classical NF-κB activity in the suppression of basal noncanonical NF-κB signaling.


Asunto(s)
Síndromes de Inmunodeficiencia/metabolismo , Leucocitos Mononucleares/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Immunoblotting , Síndromes de Inmunodeficiencia/genética , Células Jurkat , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Mutación , Subunidad p52 de NF-kappa B/metabolismo , Células 3T3 NIH , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Quinasa de Factor Nuclear kappa B
12.
EMBO Mol Med ; 6(3): 384-97, 2014 03.
Artículo en Inglés | MEDLINE | ID: mdl-24480543

RESUMEN

Type 1 interferons (IFN) protect the host against viruses by engaging a cognate receptor (consisting of IFNAR1/IFNAR2 chains) and inducing downstream signaling and gene expression. However, inflammatory stimuli can trigger IFNAR1 ubiquitination and downregulation thereby attenuating IFN effects in vitro. The significance of this paradoxical regulation is unknown. Presented here results demonstrate that inability to stimulate IFNAR1 ubiquitination in the Ifnar1(SA) knock-in mice renders them highly susceptible to numerous inflammatory syndromes including acute and chronic pancreatitis, and autoimmune and toxic hepatitis. Ifnar1(SA) mice (or their bone marrow-receiving wild type animals) display persistent immune infiltration of inflamed tissues, extensive damage and gravely inadequate tissue regeneration. Pharmacologic stimulation of IFNAR1 ubiquitination is protective against from toxic hepatitis and fulminant generalized inflammation in wild type but not Ifnar1(SA) mice. These results suggest that endogenous mechanisms that trigger IFNAR1 ubiquitination for limiting the inflammation-induced tissue damage can be purposely mimicked for therapeutic benefits.


Asunto(s)
Receptor de Interferón alfa y beta/metabolismo , Enfermedad Aguda , Animales , Trasplante de Médula Ósea , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/cirugía , Enfermedad Hepática Inducida por Sustancias y Drogas/veterinaria , Enfermedad Crónica , Femenino , Técnicas de Sustitución del Gen , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Páncreas/fisiología , Pancreatitis/inducido químicamente , Pancreatitis/patología , Pancreatitis/cirugía , Receptor de Interferón alfa y beta/genética , Regeneración , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación
13.
J Biol Chem ; 284(2): 1040-9, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-19004824

RESUMEN

We have previously shown that Fhit tumor suppressor protein interacts with Hsp60 chaperone machinery and ferredoxin reductase (Fdxr) protein. Fhit-effector interactions are associated with a Fhit-dependent increase in Fdxr stability, followed by generation of reactive oxygen species and apoptosis induction under conditions of oxidative stress. To define Fhit structural features that affect interactions, downstream signaling, and biological outcomes, we used cancer cells expressing Fhit mutants with amino acid substitutions that alter enzymatic activity, enzyme substrate binding, or phosphorylation at tyrosine 114. Gastric cancer cell clones stably expressing mutants that do not bind substrate or cannot be phosphorylated showed decreased binding to Hsp60 and Fdxr and reduced mitochondrial localization. Expression of Fhit or mutants that bind interactor proteins results in oxidative damage and accumulation of cells in G(2)/M or sub-G(1) fractions after peroxide treatment; noninteracting mutants are defective in these biological effects. Gastric cancer clones expressing noncomplexing Fhit mutants show reduction of Fhit tumor suppressor activity, confirming that substrate binding, interaction with heat shock proteins, mitochondrial localization, and interaction with Fdxr are important for Fhit tumor suppressor function.


Asunto(s)
Ácido Anhídrido Hidrolasas/química , Ácido Anhídrido Hidrolasas/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Ácido Anhídrido Hidrolasas/genética , Animales , Ciclo Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Mutación/genética , Proteínas de Neoplasias/genética , Unión Proteica , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Science ; 319(5862): 476-81, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18218901

RESUMEN

T cell receptor (TCR) and costimulatory receptor (CD28) signals cooperate in activating T cells, although understanding of how these pathways are themselves regulated is incomplete. We found that Homer2 and Homer3, members of the Homer family of cytoplasmic scaffolding proteins, are negative regulators of T cell activation. This is achieved through binding of nuclear factor of activated T cells (NFAT) and by competing with calcineurin. Homer-NFAT binding was also antagonized by active serine-threonine kinase AKT, thereby enhancing TCR signaling via calcineurin-dependent dephosphorylation of NFAT. This corresponded with changes in cytokine expression and an increase in effector-memory T cell populations in Homer-deficient mice, which also developed autoimmune-like pathology. These results demonstrate a further means by which costimulatory signals are regulated to control self-reactivity.


Asunto(s)
Proteínas Portadoras/metabolismo , Activación de Linfocitos , Factores de Transcripción NFATC/metabolismo , Linfocitos T/inmunología , Animales , Antígenos CD28/inmunología , Complejo CD3/inmunología , Calcineurina/metabolismo , Calcio/metabolismo , Proteínas Portadoras/química , Línea Celular , Células Cultivadas , Cristalografía por Rayos X , Proteínas de Andamiaje Homer , Humanos , Células Jurkat , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/química , Fosforilación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Linfocitos T/metabolismo
15.
Int J Cancer ; 120(1): 24-31, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17019711

RESUMEN

Epigenetic changes involved in cancer development, unlike genetic changes, are reversible. DNA methyltransferase and histone deacetylase inhibitors show antiproliferative effects in vitro, through tumor suppressor reactivation and induction of apoptosis. Such inhibitors have shown activity in the treatment of hematologic disorders but there is little data concerning their effectiveness in treatment of solid tumors. FHIT, WWOX and other tumor suppressor genes are frequently epigenetically inactivated in lung cancers. Lung cancer cell clones carrying conditional FHIT or WWOX transgenes showed significant suppression of xenograft tumor growth after induction of expression of the FHIT or WWOX transgene, suggesting that treatments to restore endogenous Fhit and Wwox expression in lung cancers would result in decreased tumorigenicity. H1299 lung cancer cells, lacking Fhit, Wwox, p16(INK4a) and Rassf1a expression due to epigenetic modifications, were used to assess efficacy of epigenetically targeted protocols in suppressing growth of lung tumors, by injection of 5-aza-2-deoxycytidine (AZA) and trichostatin A (TSA) in nude mice with established H1299 tumors. High doses of intraperitoneal AZA/TSA suppressed growth of small tumors but did not affect large tumors (200 mm(3)); lower AZA doses, administered intraperitoneally or intratumorally, suppressed growth of small tumors without apparent toxicity. Responding tumors showed restoration of Fhit, Wwox, p16(INKa), Rassf1a expression, low mitotic activity, high apoptotic fraction and activation of caspase 3. These preclinical studies show the therapeutic potential of restoration of tumor suppressor expression through epigenetic modulation and the promise of re-expressed tumor suppressors as markers and effectors of the responses.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Metilación de ADN , Neoplasias Pulmonares/prevención & control , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/genética , Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/metabolismo , Animales , Apoptosis , Azacitidina/análogos & derivados , Azacitidina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasas/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Transgenes , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW
16.
J Biol Chem ; 281(38): 28244-53, 2006 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-16864578

RESUMEN

The "Rosetta Stone" hypothesis proposes that the existence of a fusion protein in some organisms predicts that the separate polypeptides function in the same biochemical pathway in other organisms and may physically interact. In Drosophila melanogaster and Caenorhabditis elegans, NitFhit protein is composed of two domains, a fragile histidine triad homolog and a bacterial and plant nitrilase homolog. We assessed the biological effects of mammalian Nit1 expression in comparison with Fhit and observed that: 1) Nit1 expression was observed in most normal tissues and overlapped partially with Fhit expression; 2) Nit1-deficient mouse kidney cells exhibited accelerated proliferation, resistance to DNA damage stress, and increased cyclin D1 expression; 3) cyclin D1 was up-regulated in Nit1 null mammary gland and skin; 4) Nit1 overexpression induced caspase-dependent apoptosis in vitro; and 5) Nit1 allele deficiency led to increased incidence of N-nitrosomethylbenzylamine-induced murine forestomach tumors. Thus, the biological effects of Nit1 expression are similar to Fhit effects. Adenoviruses carrying recombinant NIT1 and FHIT induced apoptosis in Fhit- and Nit1-deficient cells, respectively, suggesting that Nit1-Fhit interaction is not essential for function of either protein. The results suggest that Nit1 and Fhit share tumor suppressor signaling pathways, while localization of the NIT1 gene at a stable, rather than fragile, chromosome site explains the paucity of gene alterations and in frequent loss of expression of the NIT1 gene in human malignancies.


Asunto(s)
Ácido Anhídrido Hidrolasas/fisiología , Aminohidrolasas/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Aminohidrolasas/genética , Animales , Apoptosis , Dominio Catalítico , Ciclina D1/análisis , Daño del ADN , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/fisiología , Neoplasias Gástricas/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA