Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Clin Cancer Res ; 25(18): 5638-5649, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308061

RESUMEN

PURPOSE: Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS.Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems. RESULTS: We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase. CONCLUSIONS: LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mutación con Pérdida de Función , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Transducción de Señal/efectos de los fármacos , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos , Síndromes Mielodisplásicos/patología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo
2.
Cancer Discov ; 9(6): 778-795, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30944118

RESUMEN

Even though the Ten-eleven translocation (TET) enzymes catalyze the generation of 5-hydroxymethylcytosines required for lineage commitment and subsequent differentiation of stem cells into erythroid cells, the mechanisms that link extracellular signals to TET activation and DNA hydroxymethylation are unknown. We demonstrate that hematopoietic cytokines phosphorylate TET2, leading to its activation in erythroid progenitors. Specifically, cytokine receptor-associated JAK2 phosphorylates TET2 at tyrosines 1939 and 1964. Phosphorylated TET2 interacts with the erythroid transcription factor KLF1, and this interaction with TET2 is increased upon exposure to erythropoietin. The activating JAK2V617F mutation seen in myeloproliferative disease patient samples and in mouse models is associated with increased TET activity and cytosine hydroxymethylation as well as genome-wide loss of cytosine methylation. These epigenetic and functional changes are also associated with increased expression of several oncogenic transcripts. Thus, we demonstrate that JAK2-mediated TET2 phosphorylation provides a mechanistic link between extracellular signals and epigenetic changes during hematopoiesis. SIGNIFICANCE: Identification of TET2 phosphorylation and activation by cytokine-stimulated JAK2 links extracellular signals to chromatin remodeling during hematopoietic differentiation. This provides potential avenues to regulate TET2 function in the context of myeloproliferative disorders and myelodysplastic syndromes associated with the JAK2V617F-activating mutation.This article is highlighted in the In This Issue feature, p. 681.


Asunto(s)
Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Janus Quinasa 2/metabolismo , Proteínas Proto-Oncogénicas/genética , Activación Transcripcional , Biomarcadores , Dioxigenasas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Fosforilación
3.
PLoS One ; 9(3): e90171, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599075

RESUMEN

The phosphoinositol-3 kinase (PI3K) pathway is highly dysregulated in squamous cell carcinoma of the head and neck (SCCHN). While inhibitors of the PI3K/AKT pathway are being developed in cancer, their efficacy does not appear to be related to the presence of mutations or amplification in pathway genes. The PI3K pathway is a major regulator of macro-autophagy, an evolutionarily conserved catabolic process that degrades cellular materials to promote cellular homeostasis and survival under stress. Employing a panel of SCCHN cell lines, we observed a significant correlation between the activity of PI3K/AKT inhibitors and their ability to induce autophagy. More specifically, resistance to these inhibitors was associated with accumulation of p62/SQSTM1, a pleotropic protein that is consumed during autophagy, while loss of autophagy was, for the first time, found to be due to silencing of an essential autophagy gene, ATG7. Moreover, modulating ATG7 and p62/SQSTM1 could regulate sensitivity to PI3K/AKT inhibitors, underscoring a mechanistic link between autophagy and drug sensitivity. Analysis of human tissues revealed progressive accumulation of p62/SQSTM1 in a significant proportion of cancer samples compared to normal tissue, suggesting that defective autophagy has relevance to SCCHN. These findings are further validated by analysis of TCGA data confirming homozygous deletion and mRNA down-regulation of ATG7 in 10.0% of SCCHN samples. Taken together, these data indicate that p62/SQSTM1 levels modulate sensitivity to PI3K/AKT inhibitors; cancers vary in their capacity to undergo autophagy through epigenetic modification and, when deficient, accumulate p62/SQSTM1; and expression of autophagy-related proteins may serve as markers for resistance to PI3K/AKT inhibitors in SCCHN.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Autofagia/efectos de los fármacos , Proteína 7 Relacionada con la Autofagia , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Resistencia a Antineoplásicos , Expresión Génica , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Mutación , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Quinoxalinas/farmacología , Proteína Sequestosoma-1 , Sulfonamidas/farmacología , Análisis de Matrices Tisulares , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo
4.
EMBO J ; 30(21): 4500-14, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21873975

RESUMEN

Tumour metastasis suppressors are inhibitors of metastasis but their mechanisms of action are generally not understood. We previously showed that the suppressor Raf kinase inhibitory protein (RKIP) inhibits breast tumour metastasis in part via let-7. Here, we demonstrate an integrated approach combining statistical analysis of breast tumour gene expression data and experimental validation to extend the signalling pathway for RKIP. We show that RKIP inhibits let-7 targets (HMGA2, BACH1) that in turn upregulate bone metastasis genes (MMP1, OPN, CXCR4). Our results reveal BACH1 as a novel let-7-regulated transcription factor that induces matrix metalloproteinase1 (MMP1) expression and promotes metastasis. An RKIP pathway metastasis signature (designated RPMS) derived from the complete signalling cascade predicts high metastatic risk better than the individual genes. These results highlight a powerful approach for identifying signalling pathways downstream of a key metastasis suppressor and indicate that analysis of genes in the context of their signalling environment is critical for understanding their predictive and therapeutic potential.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Carcinoma/diagnóstico , Carcinoma/genética , MicroARNs/fisiología , Proteínas de Unión a Fosfatidiletanolamina/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/fisiología , Neoplasias de la Mama/patología , Carcinoma/patología , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Análisis por Micromatrices , Modelos Biológicos , Metástasis de la Neoplasia , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Pronóstico , Transducción de Señal/genética , Transducción de Señal/fisiología
5.
Head Neck ; 33(12): 1774-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21438065

RESUMEN

BACKGROUND: Rapamycin is an mTOR inhibitor with preclinical efficacy in squamous cell carcinoma of the head and neck (SCCHN). However, mTOR inhibitors also increase Akt activity in SCCHN cell lines, which would promote survival and oncogenesis. Enzastaurin is an AGC kinase inhibitor with nanomolar inhibitory concentrations for Akt and protein kinase C (PKC). Moreover, Akt and PKC inhibitors have demonstrated efficacy in SCCHN. METHODS: We hypothesized that the combination of rapamycin and enzastaurin would be more effective than either agent alone. RESULTS: Rapamycin and enzastaurin generally inhibited putative targets in SCCHN cell lines in culture. In mice xenografted with CAL27 cells, rapamycin and enzastaurin produced growth delay. In contrast, the combination of rapamycin and enzastaurin caused regression of CAL27 tumors with evidence of inhibition of putative targets, survival, angiogenesis and proliferation. CONCLUSION: These data demonstrate that the combination of rapamycin and enzastaurin disrupts critical oncogenic pathways in SCCHN and has efficacy in preclinical models.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Indoles/farmacología , Sirolimus/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Neoplasias de Cabeza y Cuello/patología , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica , Inhibidores de Proteínas Quinasas/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
6.
Mol Cancer Ther ; 9(10): 2814-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20876745

RESUMEN

The number of targeted small molecules being developed in oncology is increasing rapidly. Many of these are designed to inhibit multiple kinases, and thus the mechanisms of responsiveness and predictive biomarkers can be difficult to discern. In fact, with few exceptions, multi-kinase inhibitors are developed with limited mechanism-based patient selection. Enzastaurin is a multi-kinase inhibitor being studied in several malignancies that we hypothesized would be active in squamous cell carcinoma of the head and neck, because it inhibits classic and novel protein kinase C isoforms. Indeed, enzastaurin reduced the growth of SQ-20B and CAL27 tumor xenografts, decreased proliferation in these cell lines, inhibited putative target phosphorylation, and induced cell cycle arrest. Gene expression arrays confirmed that expression of cell cycle genes, including cyclins D and E, were significantly altered by exposure to enzastaurin. However, testing a panel of squamous cell carcinoma of the head and neck cell lines revealed variable sensitivity to enzastaurin, which correlated significantly with baseline cyclin D1 protein expression. Moreover, sensitivity and resistance could be reversed, respectively, by expression or depletion of cyclin D1. Furthermore, analysis of sensitive and resistant cell lines revealed distinct differences in cyclin D1 regulation. Enzastaurin modulated cyclin D1 synthesis through an Akt-regulated pathway in the former, whereas high-level CCND1 gene amplification was present in the latter. These results underscore the critical relevance of cellular signaling context in developing cancer therapies in general and suggest that enzastaurin in particular would be most effective in tumors where baseline cyclin D1 expression is low to moderate and physiologically regulated.


Asunto(s)
Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Secuencia de Bases , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Ciclo Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Cartilla de ADN , Femenino , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Trasplante Heterólogo
7.
FASEB J ; 23(11): 3734-42, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19584300

RESUMEN

The intermediate filament (IF) protein nestin coassembles with vimentin and promotes the disassembly of these copolymers when vimentin is hyperphosphorylated during mitosis. The aim of this study is to determine the function of these nonfilamentous particles by identifying their interacting partners. In this study, we report that these disassembled vimentin/nestin complexes interact with insulin degrading enzyme (IDE). Both vimentin and nestin interact with IDE in vitro, but vimentin binds IDE with a higher affinity than nestin. Although the interaction between vimentin and IDE is enhanced by vimentin phosphorylation at Ser-55, the interaction between nestin and IDE is phosphorylation independent. Further analyses show that phosphorylated vimentin plays the dominant role in targeting IDE to the vimentin/nestin particles in vivo, while the requirement for nestin is related to its ability to promote vimentin IF disassembly. The binding of IDE to either nestin or phosphorylated vimentin regulates IDE activity differently, depending on the substrate. The insulin degradation activity of IDE is suppressed approximately 50% by either nestin or phosphorylated vimentin, while the cleavage of bradykinin-mimetic peptide by IDE is increased 2- to 3-fold. Taken together, our data demonstrate that the nestin-mediated disassembly of vimentin IFs generates a structure capable of sequestering and modulating the activity of IDE.


Asunto(s)
Insulisina/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vimentina/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Filamentos Intermedios/metabolismo , Nestina , Fosforilación , Ratas
8.
Cancer Res ; 69(1): 65-74, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19117988

RESUMEN

Protein kinase Calpha (PKCalpha) has been implicated in cancer, but the mechanism is largely unknown. Here, we show that PKCalpha promotes head and neck squamous cell carcinoma (SCCHN) by a feed-forward network leading to cell cycle deregulation. PKCalpha inhibitors decrease proliferation in SCCHN cell lines and xenografted tumors. PKCalpha inhibition or depletion in tumor cells decreases DNA synthesis by suppressing extracellular signal-regulated kinase phosphorylation and cyclin E synthesis. Additionally, PKCalpha down-regulates miR-15a, a microRNA that directly inhibits protein synthesis of cyclin E, as well as other cell cycle regulators. Furthermore, both PKCalpha and cyclin E protein expression are increased in primary tumors, and PKCalpha inversely correlates with miR-15a expression in primary tumors. Finally, PKCalpha is associated with poor prognosis in SCCHN. These results identify PKCalpha as a key regulator of SCCHN tumor cell growth by a mechanism involving activation of mitogen-activated protein kinase, an initiator of the cell cycle, and suppression of miR-15a, an inhibitor of DNA synthesis. Although the specific components may be different, this type of feed-forward loop network, consisting of a stimulus that activates a positive signal and removes a negative brake, is likely to be a general one that enables induction of DNA synthesis by a variety of growth or oncogenic stimuli.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , MicroARNs/metabolismo , Proteína Quinasa C-alfa/metabolismo , Animales , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Procesos de Crecimiento Celular/fisiología , Ciclina E/biosíntesis , ADN de Neoplasias/biosíntesis , Femenino , Expresión Génica , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/genética , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Mucosa Bucal/citología , Mucosa Bucal/enzimología , Mucosa Bucal/metabolismo , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/biosíntesis , Proteína Quinasa C-alfa/genética , Transducción de Señal
9.
Biol Chem ; 389(11): 1441-5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18783335

RESUMEN

Immunohistochemical evidence of ubiquitous distribution of the metalloprotease insulin-degrading enzyme (IDE; insulysin) in human non-malignant tissues and tumor cells is presented. Immunohistochemical staining was performed on a multi-organ tissue microarray (pancreas, lung, kidney, central/peripheral nervous system, liver, breast, placenta, myocardium, striated muscle, bone marrow, thymus, and spleen) and on a cell microarray of 31 tumor cell lines of different origin, as well as trophoblast cells and normal blood lymphocytes and granulocytes. IDE protein was expressed in all the tissues assessed and all the tumor cell lines except for Raji and HL-60. Trophoblast cells and granulocytes, but not normal lymphocytes, were also IDE-positive.


Asunto(s)
Insulisina/inmunología , Insulisina/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Insulisina/análisis , Insulisina/sangre , Especificidad de Órganos
10.
Int J Mol Med ; 22(4): 421-31, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18813847

RESUMEN

Insulin-degrading enzyme (IDE, insulysin, insulinase; EC 3.4.22.11), a thiol metalloendopeptidase, is involved in intracellular degradation of insulin, thereby inhibiting its translocation and accumulation to the nucleus. Recently, protein expression of IDE has been demonstrated in the epithelial ducts of normal breast and breast cancer tissue. Utilizing four different antibodies generated against different epitopes of the IDE molecule, we performed Western blot analysis and immunohistochemical staining on several normal human tissues, on a plethora of tumor cell lines of different tissue origin, and on malignant breast and ovarian tissue. Applying the four IDE-directed antibodies, we demonstrated IDE expression at the protein level, by means of immunoblotting and immunocytochemistry, in each of the tumor cell lines analyzed. Insulin-degrading enzyme protein expression was found in normal tissues of the kidney, liver, lung, brain, breast and skeletal muscle, as well as in breast and ovarian cancer tissues. Immunohistochemical visualization of IDE indicated cytoplasmic localization of IDE in each of the cell lines and tissues assessed. In conclusion, we performed for the first time a wide-ranging survey on IDE protein expression in normal and malignant tissues and cells thus extending our knowledge on the cellular and tissue distribution of IDE, an enzyme which to date has mainly been studied in connection with Alzheimer's disease and diabetes but not in cancer.


Asunto(s)
Insulina/metabolismo , Insulisina/metabolismo , Neoplasias/enzimología , Adulto , Anticuerpos/inmunología , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Epítopos/inmunología , Femenino , Humanos , Inmunohistoquímica , Neoplasias/patología , Especificidad de Órganos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Adhesión en Parafina , Proteínas Recombinantes de Fusión/metabolismo , Extractos de Tejidos
11.
Int J Oncol ; 30(1): 73-80, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143514

RESUMEN

Insulin is a hormone crucial to metabolism and an essential growth factor for normal and neoplastic tissues. We have now determined insulin in extracts of 23 primary breast cancer specimens and of non-neoplastic breast tissues by a chemiluminescent immunoassay. Remarkably, insulin was measured only in grade 3 tumors, whereas grade 2 carcinomas and the normal mammary gland were each insulin-negative. We also performed immunohistochemistry for insulin-degrading enzyme (IDE), a cytoplasmic zinc metalloprotease belonging to the inverzincin family and participating in insulin cleavage. IDE was detected in most insulin-positive grade 3 carcinomas, indicating that it might be dysfunctional in these anaplastic tumors. IDE was equally present in the insulin-negative grade 2 carcinomas. Moreover, five grade 3 carcinomas and one grade 2 carcinoma displayed a loss of heterozygosity in the 10q chromosomal region harboring the IDE gene, but, despite these alterations, IDE was detected immunohistochemically, indicating a retention of the second allele. Compared to the expression of IDE in 92% of the tumors examined, only 57% of 21 normal breast specimens stained positively for IDE. In contrast to this increase in IDE-positive epithelial cells in breast cancer vs. normal breast, additional immunohistochemical analysis of 17 node-positive breast carcinomas and corresponding tumor-bearing lymph nodes showed that IDE expression decreases from primary tumor to lymph node metastasis. Altogether, this study represents the first demonstration of IDE in normal and neoplastic human mammary tissues. Our present report should also provide an experimental starting point towards exploring a potential role of IDE in the control of tumor progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Mama/enzimología , Insulina/metabolismo , Insulisina/metabolismo , Adulto , Anciano , Femenino , Humanos , Inmunohistoquímica , Menopausia , Persona de Mediana Edad , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Valores de Referencia
12.
Biochem Biophys Res Commun ; 343(4): 1032-7, 2006 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-16574064

RESUMEN

Insulin degrading enzyme (IDE), a zinc metalloprotease, can specifically recognize and degrade insulin, as well as several amyloidogenic peptides such as amyloid beta (Abeta) and amylin. The disruption of IDE function in rodents leads to glucose intolerance and cerebral Abeta accumulation, hallmarks of type 2 diabetes and Alzheimer's disease, respectively. Using limited proteolysis, we found that human IDE (113kDa) can be subdivided into two roughly equal sized domains, IDE-N and IDE-C. Oligomerization plays a key role in the activity of IDE. Size-exclusion chromatography and sedimentation velocity experiments indicate that IDE-N is a monomer and IDE-C serves to oligomerize IDE-N. IDE-C alone does not have catalytic activity. It is IDE-N that contains the crucial catalytic residues, however IDE-N alone has only 2% of the catalytic activity of wild type IDE. By complexing IDE-C with IDE-N, the activity of IDE-N can be restored to approximately 30% that of wild type IDE. Fluorescence polarization assays using labeled insulin reveal that IDE-N has reduced affinity to insulin relative to wild type IDE. Together, our data reveal the modular nature of IDE. IDE-N is the catalytic domain and IDE-C facilitates substrate recognition as well as plays a key role in the oligomerization of IDE.


Asunto(s)
Insulisina/química , Dimerización , Humanos , Hidrólisis , Estructura Terciaria de Proteína , Especificidad por Sustrato
13.
J Biol Chem ; 279(22): 23073-81, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15033983

RESUMEN

ERK7 is a unique member of the extracellular signal-regulated kinase (ERK) subfamily of MAP kinases. Although ERK7 shares a TEY motif in the activation loop of the kinase, it displays constitutive activation, nuclear localization, and growth inhibitory properties that are regulated by its C-terminal domain. Because ERK7 is expressed at low levels compared with ERK2 and its activity is dependent upon its expression level, we investigated the mechanism by which ERK7 expression is regulated. We now show that ERK7 expression is regulated by ubiquitination and rapid proteosomal turnover. Furthermore, both the kinase domain and the C-terminal tail are independently degraded at a rate comparable with that of the intact protein. Analysis of a series of chimeras between ERK2 and ERK7 reveal that the N-terminal 20 amino acids of the kinase domain are a primary determinant of ERK7 degradation. Fusion of the N-terminal 20 amino acids is both necessary and sufficient to cause proteolytic degradation of both ERK2 and green fluorescent protein. Finally, ERK7 is stabilized by an N-terminal mutant of Cullin-1 suggesting that ERK7 is ubiquitinated by the Skip1-Cullin-F box complex. These results indicate that ERK7 is a highly regulated enzyme whose cellular expression and kinase activation level is tightly controlled by the ubiquitin-proteosome pathway.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Complejos Multienzimáticos/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Activación Enzimática , Humanos , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Fosforilación , Complejo de la Endopetidasa Proteasomal , Ratas , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA