Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circulation ; 144(17): 1409-1428, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34694888

RESUMEN

BACKGROUND: Complex molecular programs in specific cell lineages govern human heart development. Hypoplastic left heart syndrome (HLHS) is the most common and severe manifestation within the spectrum of left ventricular outflow tract obstruction defects occurring in association with ventricular hypoplasia. The pathogenesis of HLHS is unknown, but hemodynamic disturbances are assumed to play a prominent role. METHODS: To identify perturbations in gene programs controlling ventricular muscle lineage development in HLHS, we performed whole-exome sequencing of 87 HLHS parent-offspring trios, nuclear transcriptomics of cardiomyocytes from ventricles of 4 patients with HLHS and 15 controls at different stages of heart development, single cell RNA sequencing, and 3D modeling in induced pluripotent stem cells from 3 patients with HLHS and 3 controls. RESULTS: Gene set enrichment and protein network analyses of damaging de novo mutations and dysregulated genes from ventricles of patients with HLHS suggested alterations in specific gene programs and cellular processes critical during fetal ventricular cardiogenesis, including cell cycle and cardiomyocyte maturation. Single-cell and 3D modeling with induced pluripotent stem cells demonstrated intrinsic defects in the cell cycle/unfolded protein response/autophagy hub resulting in disrupted differentiation of early cardiac progenitor lineages leading to defective cardiomyocyte subtype differentiation/maturation in HLHS. Premature cell cycle exit of ventricular cardiomyocytes from patients with HLHS prevented normal tissue responses to developmental signals for growth, leading to multinucleation/polyploidy, accumulation of DNA damage, and exacerbated apoptosis, all potential drivers of left ventricular hypoplasia in absence of hemodynamic cues. CONCLUSIONS: Our results highlight that despite genetic heterogeneity in HLHS, many mutations converge on sequential cellular processes primarily driving cardiac myogenesis, suggesting novel therapeutic approaches.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico/genética , Organogénesis/genética , Heterogeneidad Genética , Humanos
2.
J Clin Invest ; 131(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33201861

RESUMEN

Genetic factors undoubtedly affect the development of congenital heart disease (CHD) but still remain ill defined. We sought to identify genetic risk factors associated with CHD and to accomplish a functional analysis of SNP-carrying genes. We performed a genome-wide association study (GWAS) of 4034 White patients with CHD and 8486 healthy controls. One SNP on chromosome 5q22.2 reached genome-wide significance across all CHD phenotypes and was also indicative for septal defects. One region on chromosome 20p12.1 pointing to the MACROD2 locus identified 4 highly significant SNPs in patients with transposition of the great arteries (TGA). Three highly significant risk variants on chromosome 17q21.32 within the GOSR2 locus were detected in patients with anomalies of thoracic arteries and veins (ATAV). Genetic variants associated with ATAV are suggested to influence the expression of WNT3, and the variant rs870142 related to septal defects is proposed to influence the expression of MSX1. We analyzed the expression of all 4 genes during cardiac differentiation of human and murine induced pluripotent stem cells in vitro and by single-cell RNA-Seq analyses of developing murine and human hearts. Our data show that MACROD2, GOSR2, WNT3, and MSX1 play an essential functional role in heart development at the embryonic and newborn stages.


Asunto(s)
Sitios Genéticos , Cardiopatías Congénitas/genética , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Animales , Femenino , Estudio de Asociación del Genoma Completo , Alemania/epidemiología , Cardiopatías Congénitas/epidemiología , Humanos , Masculino , Ratones , Factores de Riesgo
3.
Proc Natl Acad Sci U S A ; 117(15): 8503-8514, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32234784

RESUMEN

The specific interaction of importins with nuclear localization signals (NLSs) of cargo proteins not only mediates nuclear import but also, prevents their aberrant phase separation and stress granule recruitment in the cytoplasm. The importin Transportin-1 (TNPO1) plays a key role in the (patho-)physiology of both processes. Here, we report that both TNPO1 and Transportin-3 (TNPO3) recognize two nonclassical NLSs within the cold-inducible RNA-binding protein (CIRBP). Our biophysical investigations show that TNPO1 recognizes an arginine-glycine(-glycine) (RG/RGG)-rich region, whereas TNPO3 recognizes a region rich in arginine-serine-tyrosine (RSY) residues. These interactions regulate nuclear localization, phase separation, and stress granule recruitment of CIRBP in cells. The presence of both RG/RGG and RSY regions in numerous other RNA-binding proteins suggests that the interaction of TNPO1 and TNPO3 with these nonclassical NLSs may regulate the formation of membraneless organelles and subcellular localization of numerous proteins.


Asunto(s)
Núcleo Celular/metabolismo , Señales de Localización Nuclear , Fragmentos de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , beta Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Arginina/química , Arginina/metabolismo , Citoplasma/metabolismo , Glicina/química , Glicina/metabolismo , Células HeLa , Humanos , Fragmentos de Péptidos/química , Unión Proteica , Conformación Proteica , Proteínas de Unión al ARN/química , Serina/química , Serina/metabolismo , Tirosina/química , Tirosina/metabolismo , beta Carioferinas/química
4.
Brain Pathol ; 30(1): 36-45, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31099449

RESUMEN

Aggregation of amyloid-ß (Aß) that leads to the formation of plaques in Alzheimer's disease (AD) occurs through the stepwise formation of oligomers and fibrils. An earlier onset of aggregation is obtained upon intracerebral injection of Aß-containing brain homogenate into human APP transgenic mice that follows a prion-like seeding mechanism. Immunoprecipitation of these brain extracts with anti-Aß oligomer antibodies or passive immunization of the recipient animals abrogated the observed seeding activity, although induced Aß deposition was still evident. Here, we establish that, together with Aß monomers, Aß oligomers trigger the initial phase of Aß seeding and that the depletion of oligomeric Aß delays the aggregation process, leading to a transient reduction of seed-induced Aß deposits. This work extends the current knowledge about the role of Aß oligomers beyond its cytotoxic nature by pointing to a role in the initiation of Aß aggregation in vivo. We conclude that Aß oligomers are important for the early initiation phase of the seeding process.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Amiloide/metabolismo , Péptidos beta-Amiloides/fisiología , Animales , Encéfalo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/fisiopatología , Agregación Patológica de Proteínas/metabolismo
5.
Life Sci Alliance ; 1(5): e201800178, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30456387

RESUMEN

Systematic analysis of human arginine methylation identifies two distinct signaling modes; either isolated modifications akin to canonical post-translational modification regulation, or clustered arrays within disordered protein sequence. Hundreds of proteins contain these methyl-arginine arrays and are more prone to accumulate mutations and more tightly expression-regulated than dispersed methylation targets. Arginines within an array in the highly methylated RNA-binding protein synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP) were experimentally shown to function in concert, providing a tunable protein interaction interface. Quantitative immunoprecipitation assays defined two distinct cumulative binding mechanisms operating across 18 proximal arginine-glycine (RG) motifs in SYNCRIP. Functional binding to the methyltransferase PRMT1 was promoted by continual arginine stretches, whereas interaction with the methyl-binding protein SMN1 was arginine content-dependent irrespective of linear position within the unstructured region. This study highlights how highly repetitive modifiable amino acid arrays in low structural complexity regions can provide regulatory platforms, with SYNCRIP as an extreme example how arginine methylation leverages these disordered sequences to mediate cellular interactions.

6.
Sci Rep ; 8(1): 7084, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29728564

RESUMEN

TDP-43 and FUS are nuclear proteins with multiple functions in mRNA processing. They play key roles in ALS (amyotrophic lateral sclerosis) and FTD (frontotemporal dementia), where they are partially lost from the nucleus and aggregate in the cytoplasm of neurons and glial cells. Defects in nucleocytoplasmic transport contribute to this pathology, hence nuclear import of both proteins has been studied in detail. However, their nuclear export routes remain poorly characterized and it is unclear whether aberrant nuclear export contributes to TDP-43 or FUS pathology. Here we show that predicted nuclear export signals in TDP-43 and FUS are non-functional and that both proteins are exported independently of the export receptor CRM1/Exportin-1. Silencing of Exportin-5 or the mRNA export factor Aly/REF, as well as mutations that abrogate RNA-binding do not impair export of TDP-43 and FUS. However, artificially enlarging TDP-43 or FUS impairs their nuclear egress, suggesting that they could leave the nucleus by passive diffusion. Finally, we found that inhibition of transcription causes accelerated nuclear egress of TDP-43, suggesting that newly synthesized RNA retains TDP-43 in the nucleus, limiting its egress into the cytoplasm. Our findings implicate reduced nuclear retention as a possible factor contributing to mislocalization of TDP-43 in ALS/FTD.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Carioferinas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas de Unión al ADN/química , Humanos , Carioferinas/química , Unión Proteica , Señales de Clasificación de Proteína , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína FUS de Unión a ARN/química , Receptores Citoplasmáticos y Nucleares/química , Proteína Exportina 1
7.
Acta Neuropathol ; 131(4): 587-604, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26895297

RESUMEN

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , beta Carioferinas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Anticuerpos/farmacología , Arginina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Células Madre Embrionarias , Inhibidores Enzimáticos/farmacología , Femenino , Degeneración Lobar Frontotemporal/genética , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína FUS de Unión a ARN/inmunología , Ratas , beta Carioferinas/inmunología
8.
Nat Med ; 21(7): 802-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26099047

RESUMEN

Amyloid-ß (Aß) plaques and α-synuclein (α-syn)-rich Lewy bodies are the major neuropathological hallmarks of Alzheimer's disease (AD) and Parkinson's disease, respectively. An overlap of pathologies is found in most individuals with dementia with Lewy bodies (DLB) and in more than 50% of AD cases. Their brains display substantial α-syn accumulation not only in Lewy bodies, but also in dystrophic neurites decorating Aß plaques. Several studies report binding and coaggregation of Aß and α-syn, yet the precise role of α-syn in amyloid plaque formation remains elusive. Here we performed intracerebral injections of α-syn-containing preparations into amyloid precursor protein (APP) transgenic mice (expressing APP695(KM670/671NL) and PSEN1(L166P) under the control of the neuron-specific Thy-1 promoter; referred to here as 'APPPS1'). Unexpectedly, α-syn failed to cross-seed Aß plaques in vivo, but rather it inhibited plaque formation in APPPS1 mice coexpressing SNCA(A30P) (referred to here as 'APPPS1 × [A30P]aSYN' double-transgenic mice). This was accompanied by increased Aß levels in cerebrospinal fluid despite unchanged overall Aß levels. Notably, the seeding activity of Aß-containing brain homogenates was considerably reduced by α-syn, and Aß deposition was suppressed in grafted tissue from [A30P]aSYN transgenic mice. Thus, we conclude that an interaction between Aß and α-syn leads to inhibition of Aß deposition and to reduced plaque formation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/ultraestructura , Animales , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/patología , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/ultraestructura , Presenilina-1/metabolismo
9.
Acta Neuropathol ; 126(2): 179-88, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23775142

RESUMEN

Amyloid-ß (Aß) plaque deposition plays a central role in the pathogenesis of Alzheimer's disease (AD). Post-mortem analysis of plaque development in mouse models of AD revealed that plaques are initially small, but then increase in size and become more numerous with age. There is evidence that plaques can grow uniformly over time; however, a complementary hypothesis of plaque development is that small plaques cluster and grow together thereby forming larger plaques. To investigate the latter hypothesis, we studied plaque formation in APPPS1 mice using in vivo two-photon microscopy and immunohistochemical analysis. We used sequential pre- and post-mortem staining techniques to label plaques at different stages of development and to detect newly emerged plaques. Post-mortem analysis revealed that a subset (22 %) of newly formed plaques appeared very close (<40 µm) to pre-existing plaques and that many close plaques (25 %) that were initially separate merged over time to form one single large plaque. Our results suggest that small plaques can cluster together, thus forming larger plaques as a complementary mechanism to simple uniform plaque growth from a single initial plaque. This study deepens our understanding of Aß deposition and demonstrates that there are multiple mechanisms at play in plaque development.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Placa Amiloide/patología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Coloración y Etiquetado/métodos
10.
EMBO J ; 31(22): 4258-75, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-22968170

RESUMEN

Fused in sarcoma (FUS) is a nuclear protein that carries a proline-tyrosine nuclear localization signal (PY-NLS) and is imported into the nucleus via Transportin (TRN). Defects in nuclear import of FUS have been implicated in neurodegeneration, since mutations in the PY-NLS of FUS cause amyotrophic lateral sclerosis (ALS). Moreover, FUS is deposited in the cytosol in a subset of frontotemporal lobar degeneration (FTLD) patients. Here, we show that arginine methylation modulates nuclear import of FUS via a novel TRN-binding epitope. Chemical or genetic inhibition of arginine methylation restores TRN-mediated nuclear import of ALS-associated FUS mutants. The unmethylated arginine-glycine-glycine domain preceding the PY-NLS interacts with TRN and arginine methylation in this domain reduces TRN binding. Inclusions in ALS-FUS patients contain methylated FUS, while inclusions in FTLD-FUS patients are not methylated. Together with recent findings that FUS co-aggregates with two related proteins of the FET family and TRN in FTLD-FUS but not in ALS-FUS, our study provides evidence that these two diseases may be initiated by distinct pathomechanisms and implicates alterations in arginine methylation in pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Arginina/metabolismo , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Señales de Localización Nuclear/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/genética , Degeneración Lobar Frontotemporal/metabolismo , Silenciador del Gen , Células HeLa , Humanos , Carioferinas/genética , Metilación , Datos de Secuencia Molecular , Prolina/metabolismo , Unión Proteica , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína FUS de Unión a ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Tirosina/metabolismo
11.
J Biol Chem ; 285(34): 26174-81, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20538592

RESUMEN

Nicastrin and its relative Nicalin (Nicastrin-like protein) are both members of larger protein complexes, namely gamma-secretase and the Nicalin-NOMO (Nodal modulator) complex. The gamma-secretase complex, which contains Presenilin, APH-1, and PEN-2 in addition to Nicastrin, catalyzes the proteolytic cleavage of the transmembrane domain of various proteins including the beta-amyloid precursor protein and Notch. Nicalin and its binding partner NOMO form a complex that was shown to modulate Nodal signaling in developing zebrafish embryos. Because its experimentally determined native size (200-220 kDa) could not be satisfyingly explained by the molecular masses of Nicalin (60 kDa) and NOMO (130 kDa), we searched in affinity-purified complex preparations for additional components in the low molecular mass range. A approximately 22-kDa protein was isolated and identified by mass spectrometry as transmembrane protein 147 (TMEM147), a novel, highly conserved membrane protein with a putative topology similar to APH-1. Like Nicalin and NOMO, it localizes to the endoplasmic reticulum and is expressed during early zebrafish development. Overexpression and knockdown experiments in cultured cells demonstrate a close relationship between the three proteins and suggest that they are components of the same complex. We present evidence that, similar to gamma-secretase, its assembly is hierarchical starting with the formation of a Nicalin-NOMO intermediate. Nicalin appears to represent the limiting factor regulating the assembly rate by stabilizing the other two components. We conclude that TMEM147 is a novel core component of the Nicalin-NOMO complex, further emphasizing its similarity with gamma-secretase.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Animales , Retículo Endoplásmico , Humanos , Glicoproteínas de Membrana , Complejos Multiproteicos , Unión Proteica , Estabilidad Proteica , Pez Cebra , Proteínas de Pez Cebra
12.
J Cell Physiol ; 220(2): 367-75, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19334037

RESUMEN

We previously showed that integrin alphavbeta3 overexpression and engagement by its ligand vitronectin increased adhesion, motility, and proliferation of human ovarian cancer cells. In search of differentially regulated genes involved in these tumor biological events, we previously identified the integrin-linked kinase (ILK) to be under control of alphavbeta3. In the present investigation we demonstrated significantly upregulated ILK protein as a function of alphavbeta3 in two ovarian cancer cell lines, OV-MZ-6 and OVCAR-3, and proved co-localization at the surface of alphavbeta3-overexpressing cells adherent to vitronectin. Increase of ILK protein was reflected by enhanced ILK promoter activity, an effect, which we further characterized with regard to transcriptional response elements involved. Abrogation of NF-kappaB/c-rel or p53 binding augmented ILK promoter activity and preserved induction by alphavbeta3. The AP1-mutant exhibited decreased promoter activity but was also still inducible by alphavbeta3. Disruption of the two DNA consensus motifs for Ets proteins led to divergent observations: mutation of the Ets motif at promoter position -462 bp did not significantly alter promoter activity but still allowed response to alphavbeta3. In contrast, disruption of the second Ets motif at position -85 bp did not only lead to slightly diminished promoter activity but also, in that case, abrogated ILK promoter induction by alphavbeta3. Subsequent co-transfection studies with ets-1 in the presence of the second Ets motif led to additional induction of ILK promoter activity. Taken together, these data suggest that ets-1 binding to the second Ets DNA motif strongly contributes to alphavbeta3-mediated ILK upregulation. By increasing ILK as an important integrin-proximal kinase, alphavbeta3 may promote its intracellular signaling and tumor biological processes arising thereof in favor of ovarian cancer metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Integrina alfaVbeta3/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Femenino , Humanos , Integrina alfaVbeta3/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética
13.
Int J Biochem Cell Biol ; 40(12): 2746-61, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18577466

RESUMEN

Upon overexpression of integrin alphavbeta3 and its engagement by vitronectin, we previously showed enhanced adhesion, proliferation, and motility of human ovarian cancer cells. By studying differential expression of genes possibly related to these tumor biological events, we identified the epidermal growth-factor receptor (EGF-R) to be under control of alphavbeta3 expression levels. Thus in the present study we characterized alphavbeta3-dependent changes of EGF-R and found significant upregulation of its expression and activity which was reflected by prominent changes of EGF-R promoter activity. Upon disruption of DNA-binding motifs for the transcription factors p53, ETF, the repressor ETR, p50, and c-rel, respectively, we sought to identify DNA elements contributing to alphavbeta3-mediated EGF-R promoter induction. Both, the p53- and ETF-mutant, while exhibiting considerably lower EGF-R promoter activity than the wild type promoter, retained inducibility by alphavbeta3. Mutation of the repressor motif ETR, as expected, enhanced EGF-R promoter activity with a further moderate increase upon alphavbeta3 elevation. The p50-mutant displayed EGF-R promoter activity almost comparable to that of the wild type promoter with no impairment of induction by alphavbeta3. However, the activity of an EGF-R promoter mutant displaying a disrupted c-rel-binding motif did not only prominently decline, but, moreover, was not longer responsive to enhanced alphavbeta3, involving this DNA element in alphavbeta3-dependent EGF-R upregulation. Moreover, alphavbeta3 did not only increase the EGF-R but, moreover, also led to obvious co-clustering on the cancer cell surface. By studying alphavbeta3/EGF-R-effects on the focal adhesion kinase (FAK) and the mitogen activated protein kinases (MAPK) p44/42 (erk(-1)/erk(-2)), having important functions in synergistic crosstalk between integrins and growth-factor receptors, we found for both significant enhancement of expression and activity upon alphavbeta3/VN interaction and cell stimulation by EGF. Upregulation of the EGF-R by integrin alphavbeta3, both receptor molecules with a well-defined role as targets for cancer treatment, might represent an additional mechanism to adapt synergistic receptor signaling and crosstalk in response to an altered tumor cell microenvironment during ovarian cancer progression.


Asunto(s)
Receptores ErbB/metabolismo , Expresión Génica , Integrina alfaVbeta3/metabolismo , Neoplasias Ováricas/patología , Regulación hacia Arriba , Línea Celular Tumoral , Proliferación Celular , Receptores ErbB/genética , Femenino , Genes Reporteros , Humanos , Inmunohistoquímica , Integrina alfaVbeta3/genética , Luciferasas de Renilla/metabolismo , Neoplasias Ováricas/genética , Transfección , Vitronectina/metabolismo
14.
Pediatr Transplant ; 7(1): 46-52, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12581328

RESUMEN

We tested blood samples of 25 pediatric renal transplant recipients for Epstein-Barr virus (EBV) DNA load by quantitative polymerase chain reaction (PCR). Eleven of these transplant recipients showed clinical persistent mononucleosis-like symptoms years after transplantation (Tx). A quantitation of EBV DNA by PCR in peripheral blood lymphocyte (PBL) and serum samples revealed variable EBV DNA titers. The majority of EBV PCR results in samples of the 14 asymptomatic transplant recipients was repeatedly below detection limit. In contrast, patients with mononucleosis-like symptoms showed persistent EBV genome titers over a period of 6 months, ranging from 75 to 18 750 copies/10 000 PBL and from 680 to 335 000 copies/mL serum, respectively. One child suffering from this mononucleosis-like condition developed an EBV-associated Burkitt-like lymphoma 29 months after Tx. Whereas clinical and histological investigations did not indicate a post-transplant lymphoproliferative disorder (PTLD) until tumor detection, EBV titers in PBL and serum had been high for at least 8 months. We propose that pediatric transplant recipients who show both, recurrent mononucleosis-like symptoms and a sustained high EBV genome load, are at increased risk for severe EBV-related post-transplant complications.


Asunto(s)
ADN Viral/sangre , Herpesvirus Humano 4/aislamiento & purificación , Mononucleosis Infecciosa/diagnóstico , Trasplante de Riñón/efectos adversos , Leucocitos/virología , Trastornos Linfoproliferativos/etiología , Linfoma de Burkitt/diagnóstico , Linfoma de Burkitt/etiología , Niño , Preescolar , Femenino , Humanos , Mononucleosis Infecciosa/etiología , Mononucleosis Infecciosa/transmisión , Masculino , Reacción en Cadena de la Polimerasa , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...