Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Circulation ; 149(25): 1960-1979, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38752370

RESUMEN

BACKGROUND: Cardiomyocyte differentiation involves a stepwise clearance of repressors and fate-restricting regulators through the modulation of BMP (bone morphogenic protein)/Wnt-signaling pathways. However, the mechanisms and how regulatory roadblocks are removed with specific developmental signaling pathways remain unclear. METHODS: We conducted a genome-wide CRISPR screen to uncover essential regulators of cardiomyocyte specification in human embryonic stem cells using a myosin heavy chain 6 (MYH6)-GFP (green fluorescence protein) reporter system. After an independent secondary single guide ribonucleic acid validation of 25 candidates, we identified NF2 (neurofibromin 2), a moesin-ezrin-radixin like (MERLIN) tumor suppressor, as an upstream driver of early cardiomyocyte lineage specification. Independent monoclonal NF2 knockouts were generated using CRISPR-Cas9, and cell states were inferred through bulk RNA sequencing and protein expression analysis across differentiation time points. Terminal lineage differentiation was assessed by using an in vitro 2-dimensional-micropatterned gastruloid model, trilineage differentiation, and cardiomyocyte differentiation. Protein interaction and post-translation modification of NF2 with its interacting partners were assessed using site-directed mutagenesis, coimmunoprecipitation, and proximity ligation assays. RESULTS: Transcriptional regulation and trajectory inference from NF2-null cells reveal the loss of cardiomyocyte identity and the acquisition of nonmesodermal identity. Sustained elevation of early mesoderm lineage repressor SOX2 and upregulation of late anticardiac regulators CDX2 and MSX1 in NF2 knockout cells reflect a necessary role for NF2 in removing regulatory roadblocks. Furthermore, we found that NF2 and AMOT (angiomotin) cooperatively bind to YAP (yes-associated protein) during mesendoderm formation, thereby preventing YAP activation, independent of canonical MST (mammalian sterile 20-like serine-threonine protein kinase)-LATS (large tumor suppressor serine-threonine protein kinase) signaling. Mechanistically, cardiomyocyte lineage identity was rescued by wild-type and NF2 serine-518 phosphomutants, but not NF2 FERM (ezrin-radixin-meosin homology protein) domain blue-box mutants, demonstrating that the critical FERM domain-dependent formation of the AMOT-NF2-YAP scaffold complex at the adherens junction is required for early cardiomyocyte lineage differentiation. CONCLUSIONS: These results provide mechanistic insight into the essential role of NF2 during early epithelial-mesenchymal transition by sequestering the repressive effect of YAP and relieving regulatory roadblocks en route to cardiomyocytes.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Miocitos Cardíacos , Neurofibromina 2 , Humanos , Miocitos Cardíacos/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Sistemas CRISPR-Cas , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología
2.
Clin Nephrol ; 95(5): 256-272, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33605873

RESUMEN

OBJECTIVE: In this study, we trace the changes in the clinical and histological pattern of IgA nephritis (IgAN) in Singapore as it has evolved over 4 decades and compare the clinical, demographic, histological, and renal outcome of patients with IgAN from the 1st decade and the 4th decade. MATERIALS AND METHODS: This is a retrospective study of all histologically proven IgAN diagnosed between 1976 and 2018. Clinical, laboratory, and histological characteristics between the 1st and the 4th decade, including treatment which could influence the disease progression and renal outcome of these two groups, were compared. We used the Oxford classification to compare the renal biopsy changes for these 2 decades as we were able to retrieve 125 renal biopsy tissues for the 1st cohort of IgAN studied in the 1970s for the comparative study. RESULTS: The commonest clinical presentation throughout the first 3 decades was asymptomatic hematuria and proteinuria (63, 52, and 49%, respectively). In the 4th decade, nephrotic syndrome (31%) was the commonest followed by asymptomatic hematuria and proteinuria (30%), hypertension (21%), and chronic renal failure (11%). The data showed that treatment can modify the Oxford MEST - Crescent scores. Renin-angiotensin system (RAS) blockers modified the S scores, immunosuppressants modified the T and C scores, and combination therapy with RAS blockers and immunosuppressants modified the E, S, and T scores. CONCLUSION: The Oxford MEST classification offers a robust and expressive classification for early and late disease progression with respect to the development of end-stage renal disease (ESRD). E and S seem to be indices of continuing disease activity with progressive glomerulosclerosis, probably still amenable to therapy, but T was a predictive indicator for those destined for ESRD and no longer amenable to therapy.


Asunto(s)
Glomerulonefritis por IGA/complicaciones , Riñón/patología , Adulto , Progresión de la Enfermedad , Femenino , Glomerulonefritis por IGA/tratamiento farmacológico , Glomerulonefritis por IGA/patología , Hematuria/etiología , Humanos , Masculino , Persona de Mediana Edad , Síndrome Nefrótico/etiología , Proteinuria/etiología , Estudios Retrospectivos , Adulto Joven
3.
J Biol Chem ; 293(1): 163-176, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29109149

RESUMEN

Oxidative and endoplasmic reticulum (ER) stresses are hallmarks of the pathophysiology of ALS and other neurodegenerative diseases. In these stresses, different kinases phosphorylate eukaryotic initiation factor eIF2α, enabling the translation of stress response genes; among these is GADD34, the protein product of which recruits the α-isoform of protein phosphatase 1 catalytic subunit (PP1α) and eIF2α to assemble a phosphatase complex catalyzing eIF2α dephosphorylation and resumption of protein synthesis. Aberrations in this pathway underlie the aforementioned disorders. Previous observations indicating that GADD34 is induced by arsenite, a thiol-directed oxidative stressor, in the absence of eIF2α phosphorylation suggest other roles for GADD34. Here, we report that arsenite-induced oxidative stress differs from thapsigargin- or tunicamycin-induced ER stress in promoting GADD34 transcription and the preferential translation of its mRNA in the absence of eIF2α phosphorylation. Arsenite also stabilized GADD34 protein, slowing its degradation. In response to oxidative stress, but not ER stress, GADD34 recruited TDP-43, and enhanced cytoplasmic distribution and cysteine modifications of TDP-43 promoted its binding to GADD34. Arsenite also recruited a TDP-43 kinase, casein kinase-1ϵ (CK1ϵ), to GADD34. Concomitant with TDP-43 aggregation and proteolysis after prolonged arsenite exposure, GADD34-bound CK1ϵ catalyzed TDP-43 phosphorylations at serines 409/410, which were diminished or absent in GADD34-/- cells. Our findings highlight that the phosphatase regulator, GADD34, also functions as a kinase scaffold in response to chronic oxidative stress and recruits CK1ϵ and oxidized TDP-43 to facilitate its phosphorylation, as seen in TDP-43 proteinopathies.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Estrés Oxidativo/fisiología , Proteína Fosfatasa 1/metabolismo , Proteinopatías TDP-43/metabolismo , Animales , Arsenitos/farmacología , Caseína Cinasa 1 épsilon/metabolismo , Proteínas de Ciclo Celular/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Proteína Fosfatasa 1/deficiencia
4.
EMBO J ; 31(5): 1308-19, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22252131

RESUMEN

Phosphotyrosine-binding domains, typified by the SH2 (Src homology 2) and PTB domains, are critical upstream components of signal transduction pathways. The E3 ubiquitin ligase Hakai targets tyrosine-phosphorylated E-cadherin via an uncharacterized domain. In this study, the crystal structure of Hakai (amino acids 106-206) revealed that it forms an atypical, zinc-coordinated homodimer by utilizing residues from the phosphotyrosine-binding domain of two Hakai monomers. Hakai dimerization allows the formation of a phosphotyrosine-binding pocket that recognizes specific phosphorylated tyrosines and flanking acidic amino acids of Src substrates, such as E-cadherin, cortactin and DOK1. NMR and mutational analysis identified the Hakai residues required for target binding within the binding pocket, now named the HYB domain. ZNF645 also possesses a HYB domain but demonstrates different target specificities. The HYB domain is structurally different from other phosphotyrosine-binding domains and is a potential drug target due to its novel structural features.


Asunto(s)
Cadherinas/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Análisis Mutacional de ADN , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
5.
J Biol Chem ; 288(46): 33146-55, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24092754

RESUMEN

In mammalian cells, metabolic and environmental stress increases the phosphorylation of the eukaryotic translational initiation factor, eIF2α, and attenuates global protein synthesis. Subsequent transcriptional activation of GADD34 assembles an eIF2α phosphatase that feeds back to restore mRNA translation. Active proteasomal degradation of GADD34 protein then reestablishes the sensitivity of cells to subsequent bouts of stress. Mass spectrometry established GADD34 phosphorylation on multiple serines, threonines, and tyrosines. Phosphorylation at tyrosine 262 enhanced the rate of the GADD34 protein turnover. Substrate-trapping studies identified TC-PTP (PTPN2) as a potential GADD34 phosphatase, recognizing phosphotyrosine 262. Reduced GADD34 protein levels in TC-PTP-null MEFs following ER stress emphasized the importance of TC-PTP in determining the cellular levels of GADD34 protein. The susceptibility of TC-PTP-null MEFs to ER stress-induced apoptosis was significantly ameliorated by ectopic expression of GADD34. The data suggested that GADD34 phosphorylation on tyrosine 262 modulates endoplasmic reticulum stress signaling and cell fate.


Asunto(s)
Apoptosis/fisiología , Estrés del Retículo Endoplásmico/fisiología , Proteína Fosfatasa 1/metabolismo , Proteolisis , Transducción de Señal/fisiología , Animales , Células COS , Chlorocebus aethiops , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Fosforilación/fisiología , Proteína Fosfatasa 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Tirosina/genética , Tirosina/metabolismo
6.
J Biol Chem ; 286(26): 23102-12, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531714

RESUMEN

SHP2 is a tyrosine phosphatase involved in the activation of the Ras/ERK signaling pathway downstream of a number of receptor tyrosine kinases. One of the proposed mechanisms involving SHP2 in this context is to dephosphorylate and inactivate inhibitors of the Ras/ERK pathway. Two protein families bearing a unique, common domain, Sprouty and SPRED proteins, are possible candidates because they have been reported to inhibit the Ras/ERK pathway upon FGF activation. We tested whether any of these proteins are likely substrates of SHP2. Our findings indicate that Sprouty2 binds to the C-terminal tail of SHP2, which is an unlikely substrate binding site, whereas SPRED proteins bind to the tyrosine phosphatase domain that is known to be the binding site for its substrates. Overexpressed SHP2 was able to dephosphorylate SPREDs but not Sprouty2. Finally, we found two tyrosine residues on SPRED1 that are required, when phosphorylated, to inhibit Ras/ERK activation and identified Tyr-420 as a specific dephosphorylation target of SHP2. The evidence obtained indicates that SPRED1 is a likely substrate of SHP2, whose tyrosine dephosphorylation is required to attenuate the inhibitory action of SPRED1 in the Ras/ERK pathway.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Células PC12 , Unión Proteica , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Ratas , Proteínas Represoras/genética , Proteínas ras/genética , Proteínas ras/metabolismo
7.
J Biol Chem ; 285(46): 35374-85, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20736167

RESUMEN

The mammalian SPRED (Sprouty-related protein with an EVH1 domain) proteins include a family of three members, SPRED1-3. Currently, little is known about their biochemistry. The best described, SPRED1, has been shown to inhibit the Ras/ERK pathway downstream of Ras. All three SPREDs have a cysteine-rich domain (CRD) that has high homology to the CRD of the Sprouty family of proteins, several of which are also Ras/ERK inhibitors. In the belief that binding partners would clarify SPRED function, we assayed for their associated proteins. Here, we describe the direct and endogenous interaction of SPRED1 and SPRED2 with the novel kinase, DYRK1A. DYRK1A has become the subject of recent research focus as it plays a central role in Caenorhabditis elegans oocyte maturation and egg activation, and there is strong evidence that it could be involved in Down syndrome in humans. Both SPRED1 and SPRED2 inhibit the ability of DYRK1A to phosphorylate its substrates, Tau and STAT3. This inhibition occurs via an interaction of the CRD of the SPREDs with the kinase domain of DYRK1A. DYRK1A substrates must bind to the kinase to enable phosphorylation, and SPRED proteins compete for the same binding site to modify this process. Our accumulated evidence indicates that the SPRED proteins are likely physiological modifiers of DYRK1A.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Acetilación , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular , Cisteína/genética , Cisteína/metabolismo , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , Proteínas Represoras/genética , Factor de Transcripción STAT3/metabolismo , Especificidad por Sustrato , Proteína p53 Supresora de Tumor/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteínas tau/metabolismo , Quinasas DyrK
8.
Cancer Res ; 80(24): 5619-5632, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33067269

RESUMEN

A subset of Wnt-addicted cancers are sensitive to targeted therapies that block Wnt secretion or receptor engagement. RNF43 loss-of-function (LOF) mutations that increase cell surface Wnt receptor abundance cause sensitivity to Wnt inhibitors. However, it is not clear which of the clinically identified RNF43 mutations affect its function in vivo. We assayed 119 missense and 45 truncating RNF43 mutations found in human cancers using a combination of cell-based reporter assays, genome editing, flow cytometry, and immunofluorescence microscopy. Five common germline variants of RNF43 exhibited wild-type activity. Cancer-associated missense mutations in the RING ubiquitin ligase domain and a subset of mutations in the extracellular domain hyperactivate Wnt/ß-catenin signaling through formation of inactive dimers with endogenous RNF43 or ZNRF3. RNF43 C-terminal truncation mutants, including the common G659fs mutant are LOF specifically when endogenous mutations are examined, unlike their behavior in transient transfection assays. Patient-derived xenografts and cell lines with C-terminal truncations showed increased cell surface Frizzled and Wnt/ß-catenin signaling and were responsive to porcupine (PORCN) inhibition in vivo, providing clear evidence of RNF43 impairment. Our study provides potential guidelines for patient assignment, as virtually all RNF43 nonsense and frameshift mutations, including those in the C-terminal domain and a large number of patient-associated missense mutations in the RING domain and N-terminal region compromise its activity, and therefore predict response to upstream Wnt inhibitors in cancers without microsatellite instability. This study expands the landscape of actionable RNF43 mutations, extending the benefit of these therapies to additional patients. SIGNIFICANCE: Systematic examination of patient-derived RNF43 mutations identifies rules to guide patient selection, including that truncation or point mutations in well-defined functional domains sensitize cancers to PORCN inhibitors.


Asunto(s)
Mutación , Neoplasias/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Aciltransferasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Endocitosis/fisiología , Receptores Frizzled/metabolismo , Regulación Neoplásica de la Expresión Génica , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Multimerización de Proteína , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
NAR Genom Bioinform ; 2(2): lqaa013, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33575575

RESUMEN

Comprehensive understanding of aberrant splicing in gastric cancer is lacking. We RNA-sequenced 19 gastric tumor-normal pairs and identified 118 high-confidence tumor-associated (TA) alternative splicing events (ASEs) based on high-coverage sequencing and stringent filtering, and also identified 8 differentially expressed splicing factors (SFs). The TA ASEs occurred in genes primarily involved in cytoskeletal organization. We constructed a correlative network between TA ASE splicing ratios and SF expression, replicated it in independent gastric cancer data from The Cancer Genome Atlas and experimentally validated it by knockdown of the nodal SFs (PTBP1, ESRP2 and MBNL1). Each SF knockdown drove splicing alterations in several corresponding TA ASEs and led to alterations in cellular migration consistent with the role of TA ASEs in cytoskeletal organization. We have therefore established a robust network of dysregulated splicing associated with tumor invasion in gastric cancer. Our work is a resource for identifying oncogenic splice forms, SFs and splicing-generated tumor antigens as biomarkers and therapeutic targets.

10.
Cancer Res ; 66(4): 2048-58, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16489004

RESUMEN

The Sprouty proteins are increasingly being recognized to be deregulated in various types of cancers. This deregulation is often associated with aberrant signaling of receptor tyrosine kinases and its downstream effectors, leading to the mitogen-activated protein kinase (MAPK) signaling pathway. In human hepatocellular carcinoma, where the MAPK activity is enhanced via multiple hepatocarcinogenic factors, we observed a consistent reduced expression of the sprouty 2 (Spry2) transcript and protein in malignant hepatocytes compared with normal or cirrhotic hepatocytes. The expression pattern of Spry2 in hepatocellular carcinoma resembles that of several potential tumor markers of hepatocellular carcinoma and also that of several angiogenic factors and growth factor receptors. In contrast to previous studies of Spry2 down-regulation in other cancers, we have ruled out loss of heterozygosity or the methylation of promoter sites, two common mechanisms responsible for the silencing of genes with tumor suppressor properties. Functionally, we show that Spry2 inhibits both extracellular signal-regulated kinase signaling as well as proliferation in hepatocellular carcinoma cell lines, whereas knocking down Spry2 levels in NIH3T3 cells causes mild transformation. Our study clearly indicates a role for Spry2 in hepatocellular carcinoma, and an understanding of the regulatory controls of its expression could provide new means of regulating the angiogenic switch in this hypervascular tumor, thereby potentially controlling tumor growth.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metilación de ADN , Regulación hacia Abajo , Factores de Crecimiento de Fibroblastos/farmacología , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/genética , Pérdida de Heterocigocidad , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética
11.
Cell Death Differ ; 25(2): 255-267, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28984870

RESUMEN

Phosphorylation of the eukaryotic translation initiation factor, eIF2α, by stress-activated protein kinases and dephosphorylation by the growth arrest and DNA damage-inducible protein (GADD34)-containing phosphatase is a central node in the integrated stress response. Mass spectrometry demonstrated GADD34 acetylation at multiple lysines. Substituting K315 and K322 with alanines or glutamines did not impair GADD34's ability to recruit protein phosphatase 1α (PP1α) or eIF2α, suggesting that GADD34 acetylation did not modulate eIF2α phosphatase activity. Arsenite (Ars)-induced oxidative stress increased cellular GADD34 levels and enhanced Sirtuin 1 (SIRT1) recruitment to assemble a cytoplasmic complex containing GADD34, PP1α, eIF2α and SIRT1. Induction of GADD34 in WT MEFs paralleled the dephosphorylation of eIF2α (phosphoserine-51) and SIRT1 (phosphoserine-47). By comparison, eIF2α and SIRT1 were persistently phosphorylated in Ars-treated GADD34-/- MEFs. Expressing WT GADD34, but not a mutant unable to bind PP1α in GADD34-/- MEFs restored both eIF2α and SIRT1 dephosphorylation. SIRT1 dephosphorylation increased its deacetylase activity, measured in vitro and in cells. Loss of function of GADD34 or SIRT1 enhanced cellular p-eIF2α levels and attenuated cell death following Ars exposure. These results highlighted a novel role for the GADD34/PP1α complex in coordinating the dephosphorylation and reactivation of eIF2α and SIRT1 to determine cell fate following oxidative stress.


Asunto(s)
Proteína Fosfatasa 1/metabolismo , Sirtuina 1/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Estrés Oxidativo , Fosforilación , Proteína Fosfatasa 1/deficiencia , Proteína Fosfatasa 1/genética
12.
Mol Cell Biol ; 22(22): 7953-66, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12391162

RESUMEN

Sprouty (Spry) proteins have been revealed as inhibitors of the Ras/mitogen-activated protein kinase (MAPK) cascade, a pathway crucial for developmental processes initiated by activation of various receptor tyrosine kinases. In COS-1 and Swiss 3T3 cells, all Spry isoforms translocate to the plasma membrane, notably ruffles, following activation. Here we show that microinjection of active Rac induced the translocation of Spry isoforms, indicating that the target of the Spry translocation domain (SpryTD) is downstream of active Rac. Targeted disruption of actin polymerization revealed that the SpryTD target appeared upstream of cytoskeletal rearrangements. Accumulated evidence indicated that phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] is the likely SpryTD target. Human Spry2TD (hSpry2TD) binds to PtdIns(4,5)P(2) in vesicle-binding assays. hSpry2TD colocalizes with the pleckstrin homology domain of phospholipase Cdelta, which binds PtdIns(4,5)P(2). The plasma membrane localization of hSpry2TD was abolished in ionomycin-treated MDCK cells or when PtdIns(4,5)P(2) was specifically dephosphorylated by overexpression of an engineered, green fluorescent protein-tagged inositol 5-phosphatase. Similarly, Spred, a novel Ras/MAPK inhibitor recently found to contain the conserved cysteine-rich SpryTD, also translocated to peripheral membranes and bound to PtdIns(4,5)P(2). Alignment of the Spry and Spred proteins led us to identify a translocation-defective point mutant, hSpry2 D252. Targeting of hSpry2 to PtdIns(4,5)P(2) was shown to be essential for the down-regulation of Ras/MAPK signaling.


Asunto(s)
Membrana Celular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Transporte de Proteínas/fisiología , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Membrana Celular/química , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Inmunohistoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Microinyecciones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Fosfolipasa C delta , Unión Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Alineación de Secuencia , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas ras/metabolismo
13.
Cancer Lett ; 242(2): 141-50, 2006 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-16469433

RESUMEN

The Ras/Erk signaling pathway has a central role in development of multi-cellular organisms as well as in signal transmission in the mature individual. Recently, a family of genes, designated Sprouty, induced by the Ras/Erk pathway was found to specify proteins that inhibited the upstream pathway. Being in a position that is likely to control well-characterized oncogene products suggested that the expression levels of the Sprouty genes may be relevant in human carcinogenesis. Early data on the deregulation of Sprouty expression in breast, prostate and liver cancers is discussed along with the notion that some of them might have potential as tumour markers or that the derived proteins may act as tumour suppressors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/fisiología , Fosfoproteínas/fisiología , Neoplasias de la Próstata/metabolismo , Animales , Biomarcadores de Tumor , Femenino , Humanos , Masculino , Neoplasias Mamarias Animales/metabolismo , Proteínas de la Membrana/biosíntesis , Ratones , Modelos Biológicos , Fosfoproteínas/biosíntesis , Transducción de Señal , Distribución Tisular
14.
Cancer Res ; 64(17): 6127-36, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342396

RESUMEN

Sprouty (Spry) proteins were found to be endogenous inhibitors of the Ras/mitogen-activated protein kinase pathway that play an important role in the remodeling of branching tissues. We investigated Spry expression levels in various cancers and found that Spry1 and Spry2 were down-regulated consistently in breast cancers. Such prevalent patterns of down-regulation may herald the later application of these isoforms as tumor markers that are breast cancer specific and more profound than currently characterized markers. Spry1 and 2 were expressed specifically in the luminal epithelial cells of breast ducts, with higher expression during stages of tissue remodeling when the epithelial ducts are forming and branching. These findings suggest that Sprys might be involved as a modeling counterbalance and surveillance against inappropriate epithelial expansion. The abrogation of endogenous Spry activity in MCF-7 cells by the overexpression of a previously characterized dominant-negative mutant of Spry, hSpry2Y55F resulted in enhanced cell proliferation in vitro. The hSpry2Y55F stably expressing cells also formed larger and greater number of colonies in the soft-agar assay. An in vivo nude mice assay showed a dramatic increase in the tumorigenic potential of hSpry2Y55F stable cells. The consistent down-regulation of Spry1 and 2 in breast cancer and the experimental evidence using a dominant-negative hSpry2Y55F indicate that Spry proteins may actively maintain tissue integrity that runs amok when their expression is decreased below normal threshold levels. This alludes to a previously unrecognized role for Sprys in cancer development.


Asunto(s)
Neoplasias de la Mama/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/biosíntesis , Fosfoproteínas/biosíntesis , Biosíntesis de Proteínas , Proteínas Adaptadoras Transductoras de Señales , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas , Proteínas/antagonistas & inhibidores , Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo
15.
Nat Commun ; 7: 12061, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27389904

RESUMEN

Coronary flow (CF) measured ex vivo is largely determined by capillary density that reflects angiogenic vessel formation in the heart in vivo. Here we exploit this relationship and show that CF in the rat is influenced by a locus on rat chromosome 2 that is also associated with cardiac capillary density. Mitochondrial tryptophanyl-tRNA synthetase (Wars2), encoding an L53F protein variant within the ATP-binding motif, is prioritized as the candidate at the locus by integrating genomic data sets. WARS2(L53F) has low enzyme activity and inhibition of WARS2 in endothelial cells reduces angiogenesis. In the zebrafish, inhibition of wars2 results in trunk vessel deficiencies, disordered endocardial-myocardial contact and impaired heart function. Inhibition of Wars2 in the rat causes cardiac angiogenesis defects and diminished cardiac capillary density. Our data demonstrate a pro-angiogenic function for Wars2 both within and outside the heart that may have translational relevance given the association of WARS2 with common human diseases.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genoma , Células Endoteliales de la Vena Umbilical Humana/enzimología , Mitocondrias/genética , Neovascularización Fisiológica/genética , Triptófano-ARNt Ligasa/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos/química , Embrión no Mamífero , Sitios Genéticos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Mitocondrias/metabolismo , Miocardio/citología , Miocardio/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Triptófano-ARNt Ligasa/antagonistas & inhibidores , Triptófano-ARNt Ligasa/metabolismo , Pez Cebra
16.
Cell Signal ; 14(1): 11-20, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11747984

RESUMEN

The family of docker proteins containing phosphotyrosine-binding (PTB) domains appears to represent a family of critically positioned and exquisitely controlled signalling proteins that relay signals from the activated receptors to downstream pathways. These proteins all have a membrane attachment domain, a PTB domain that targets the protein to a subset of receptors and a number of phosphorylatable tyrosines that dock other signalling proteins. Evidence is accruing that suggests that the PTB domain has evolved from a pleckstrin homology (PH) domain to bind to a range of sequences that, while bestowing specificity, allows switching of the docker protein between receptors or signalling systems. The history of the PTB domain and how it influences the participation of docker protein in various signalling pathways are discussed.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Proteínas/química , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Evolución Molecular , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Estructura Terciaria de Proteína , Proteínas/fisiología , Alineación de Secuencia , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal
17.
Cell Rep ; 11(12): 1885-91, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26095357

RESUMEN

The attenuation of protein synthesis via the phosphorylation of eIF2α is a major stress response of all eukaryotic cells. The growth-arrest- and DNA-damage-induced transcript 34 (GADD34) bound to the serine/threonine protein phosphatase 1 (PP1) is the necessary eIF2α phosphatase complex that returns mammalian cells to normal protein synthesis following stress. The molecular basis by which GADD34 recruits PP1 and its substrate eIF2α are not fully understood, hindering our understanding of the remarkable selectivity of the GADD34:PP1 phosphatase for eIF2α. Here, we report detailed structural and functional analyses of the GADD34:PP1 holoenzyme and its recruitment of eIF2α. The data highlight independent interactions of PP1 and eIF2α with GADD34, demonstrating that GADD34 functions as a scaffold both in vitro and in cells. This work greatly enhances our molecular understanding of a major cellular eIF2α phosphatase and establishes the foundation for future translational work.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Proteína Fosfatasa 1/metabolismo , Relación Estructura-Actividad , Animales , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Daño del ADN/genética , Escherichia coli , Factor 2 Eucariótico de Iniciación/química , Fosforilación , Biosíntesis de Proteínas/genética , Proteína Fosfatasa 1/química
18.
J Biol Chem ; 283(3): 1679-1691, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17974561

RESUMEN

The Sprouty (Spry) proteins function as inhibitors of the Ras-ERK pathway downstream of various receptor tyrosine kinases. In this study, we have identified Tesk1 (testicular protein kinase 1) as a novel regulator of Spry2 function. Endogenous Tesk1 and Spry2 exist in a complex in cell lines and mouse tissues. Tesk1 coexpression relocalizes Spry2 to vesicles including endosomes, inhibiting its translocation to membrane ruffles upon growth factor stimulation. Independent of its kinase activity, Tesk1 binding leads to a loss of Spry2 function as an inhibitor of ERK phosphorylation and reverses inhibition of basic fibroblast growth factor (bFGF)- and nerve growth factor-induced neurite outgrowth in PC12 cells by Spry2. Furthermore, depletion of endogenous Tesk1 in PC12 cells leads to a reduction in neurite outgrowth induced by bFGF. Tesk1 nullifies the inhibitory effect of Spry2 by abrogating its interaction with the adaptor protein Grb2 and interfering with its serine dephosphorylation upon bFGF and FGF receptor 1 stimulation by impeding its binding to the catalytic subunit of protein phosphatase 2A. A construct of Tesk1 that binds to Spry2 but does not localize to the vesicles does not interfere with its function, highlighting the importance of subcellular localization of Tesk1 in this context. Conversely, Tesk1 does not affect interaction of Spry2 with the E3 ubiquitin ligase, c-Cbl, and consequently, does not affect its inhibition of Cbl-mediated ubiquitination of the epidermal growth factor receptor. By selectively modulating the downstream effects of Spry2, Tesk1 may thus serve as a molecular determinant of the signaling outcome.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Vesículas Citoplasmáticas/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Proteína Adaptadora GRB2/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Neuritas/metabolismo , Células PC12 , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ratas , Proteínas Represoras/metabolismo , Serina/metabolismo , Fracciones Subcelulares , Ubiquitinación , Proteínas ras/metabolismo
19.
J Biol Chem ; 282(12): 9117-26, 2007 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-17255109

RESUMEN

In the context of fibroblast growth factor (FGF) signaling, Sprouty2 (Spry2) is the most profound inhibitor of the Ras/ERK pathway as compared with other Spry isoforms. An exclusive, necessary, but cryptic PXXPXR motif in the C terminus of Spry2 is revealed upon stimulation. The activation of Spry2 appears to be linked to sequences in the N-terminal half of the protein and correlated with a bandshifting seen on SDS-PAGE. The band-shifting is likely caused by changes in the phosphorylation status of key Ser and Thr residues following receptor stimulation. Dephosphorylation of at least two conserved Ser residues (Ser-112 and Ser-115) within a conserved Ser/Thr sequence is accomplished upon stimulation by a phosphatase that binds to Spry2 around residues 50-60. We show that human Spry2 co-immunoprecipitates with both the catalytic and the regulatory subunits of protein phosphatase 2A (PP2A-C and PP2A-A, respectively) in cells upon FGF receptor (FGFR) activation. PP2A-A binds directly to Spry2, but not to Spry2Delta50-60 (Delta50-60), and the activity of PP2A increases with both FGF treatment and FGFR1 overexpression. c-Cbl and PP2A-A compete for binding centered around Tyr-55 on Spry2. We show that there are at least two distinct pools of Spry2, one that binds PP2A and another that binds c-Cbl. c-Cbl binding likely targets Spry2 for ubiquitin-linked destruction, whereas the phosphatase binding and activity are necessary to dephosphorylate specific Ser/Thr residues. The resulting change in tertiary structure enables the Pro-rich motif to be revealed with subsequent binding of Grb2, a necessary step for Spry2 to act as a Ras/ERK pathway inhibitor in FGF signaling.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas Fosfatasas/química , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Proteínas de la Membrana , Datos de Secuencia Molecular , Células PC12 , Fosforilación , Unión Proteica , Proteína Fosfatasa 2 , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ratas , Homología de Secuencia de Aminoácido , Transducción de Señal
20.
J Biol Chem ; 281(40): 29993-30000, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-16893902

RESUMEN

Because the Sprouty (Spry) proteins were shown to be inhibitors of the mainstream Ras/ERK pathway, there has been considerable interest in ascertaining their mechanism of action especially since a possible role as tumor suppressors for these inhibitory proteins has been suggested. We compared the ability of the mammalian Spry isoforms to inhibit the Ras/ERK pathway in the context of fibroblast growth factor receptor (FGFR) signaling. Spry2 is considerably more inhibitory than Spry1 or Spry4, and this correlates with the binding to Grb2 via a C-terminal proline-rich sequence that is found exclusively on Spry2. This PXXPXR motif binds directly to the N-terminal Src homology domain 3 of Grb2, and when added onto the C terminus of Spry4 the resultant chimera inhibits the Ras/ERK pathway. The ability to inhibit neurite outgrowth in PC-12 cells correlates with the propensity of Spry isoforms and engineered constructs to inhibit the phosphorylation of ERK1/2. The PXXPXR motif is cryptic in unstimulated cells, and it is postulated that Spry2 undergoes a conformational change following FGFR stimulation, enabling the subsequent interaction with Grb2. We present evidence that Spry2 can compete with the RasGEF (guanine nucleotide exchange factor) SOS1 for binding to Grb2, resulting in the inhibition of phosphorylation of ERK1/2.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Fragmentos de Péptidos/fisiología , Proteínas/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Proteínas ras/antagonistas & inhibidores , Dominios Homologos src/fisiología , Animales , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Células PC12 , Fosforilación , Unión Proteica , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA