Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Biomed Eng ; 8(3): 214-232, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37814006

RESUMEN

Developing therapeutic antibodies is laborious and costly. Here we report a method for antibody discovery that leverages the Illumina HiSeq platform to, within 3 days, screen in the order of 108 antibody-antigen interactions. The method, which we named 'deep screening', involves the clustering and sequencing of antibody libraries, the conversion of the DNA clusters into complementary RNA clusters covalently linked to the instrument's flow-cell surface on the same location, the in situ translation of the clusters into antibodies tethered via ribosome display, and their screening via fluorescently labelled antigens. By using deep screening, we discovered low-nanomolar nanobodies to a model antigen using 4 × 106 unique variants from yeast-display-enriched libraries, and high-picomolar single-chain antibody fragment leads for human interleukin-7 directly from unselected synthetic repertoires. We also leveraged deep screening of a library of 2.4 × 105 sequences of the third complementarity-determining region of the heavy chain of an anti-human epidermal growth factor receptor 2 (HER2) antibody as input for a large language model that generated new single-chain antibody fragment sequences with higher affinity for HER2 than those in the original library.


Asunto(s)
Anticuerpos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Anticuerpos/genética , Anticuerpos/metabolismo , Biblioteca de Genes , Fragmentos de Inmunoglobulinas , Ribosomas/genética , Ribosomas/metabolismo
2.
Front Immunol ; 14: 1107848, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936963

RESUMEN

Introduction: Humanized mice are emerging as valuable models to experimentally evaluate the impact of different immunotherapeutics on the human immune system. These immunodeficient mice are engrafted with human cells or tissues, that then mimic the human immune system, offering an alternative and potentially more predictive preclinical model. Immunodeficient NSG mice engrafted with human CD34+ cord blood stem cells develop human T cells educated against murine MHC. However, autoimmune graft versus host disease (GvHD), mediated by T cells, typically develops 1 year post engraftment. Methods: Here, we have used the development of GvHD in NSG mice, using donors with HLA alleles predisposed to autoimmunity (psoriasis) to weight in favor of GvHD, as an endpoint to evaluate the relative potency of monoclonal and BiSpecific antibodies targeting PD-1 and CTLA-4 to break immune tolerance. Results: We found that treatment with either a combination of anti-PD-1 & anti-CTLA-4 mAbs or a quadrivalent anti-PD-1/CTLA-4 BiSpecific (MEDI8500), had enhanced potency compared to treatment with anti-PD-1 or anti-CTLA-4 monotherapies, increasing T cell activity both in vitro and in vivo. This resulted in accelerated development of GvHD and shorter survival of the humanized mice in these treatment groups commensurate with their on target activity. Discussion: Our findings demonstrate the potential of humanized mouse models for preclinical evaluation of different immunotherapies and combinations, using acceleration of GvHD development as a surrogate of aggravated antigenic T-cell response against host.


Asunto(s)
Enfermedad Injerto contra Huésped , Inhibidores de Puntos de Control Inmunológico , Humanos , Animales , Ratones , Ratones SCID , Linfocitos T , Autoinmunidad
3.
MAbs ; 14(1): 2095701, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35799328

RESUMEN

Although monoclonal antibodies have greatly improved cancer therapy, they can trigger side effects due to on-target, off-tumor toxicity. Over the past decade, strategies have emerged to successfully mask the antigen-binding site of antibodies, such that they are only activated at the relevant site, for example, after proteolytic cleavage. However, the methods for designing an ideal affinity-based mask and what parameters are important are not yet well understood. Here, we undertook mechanistic studies using three masks with different properties and identified four critical factors: binding site and affinity, as well as association and dissociation rate constants, which also played an important role. HDX-MS was used to identify the location of binding sites on the antibody, which were subsequently validated by obtaining a high-resolution crystal structure for one of the mask-antibody complexes. These findings will inform future designs of optimal affinity-based masks for antibodies and other therapeutic proteins.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Monoclonales/química , Afinidad de Anticuerpos , Sitios de Unión
4.
Sci Rep ; 12(1): 3114, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210470

RESUMEN

On 11th March 2020, the UK government announced plans for the scaling of COVID-19 testing, and on 27th March 2020 it was announced that a new alliance of private sector and academic collaborative laboratories were being created to generate the testing capacity required. The Cambridge COVID-19 Testing Centre (CCTC) was established during April 2020 through collaboration between AstraZeneca, GlaxoSmithKline, and the University of Cambridge, with Charles River Laboratories joining the collaboration at the end of July 2020. The CCTC lab operation focussed on the optimised use of automation, introduction of novel technologies and process modelling to enable a testing capacity of 22,000 tests per day. Here we describe the optimisation of the laboratory process through the continued exploitation of internal performance metrics, while introducing new technologies including the Heat Inactivation of clinical samples upon receipt into the laboratory and a Direct to PCR protocol that removed the requirement for the RNA extraction step. We anticipate that these methods will have value in driving continued efficiency and effectiveness within all large scale viral diagnostic testing laboratories.


Asunto(s)
SARS-CoV-2
5.
Cancer Immunol Res ; 10(2): 200-214, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34937728

RESUMEN

Checkpoint blockade therapies targeting PD-1/PD-L1 and CTLA-4 are clinically successful but also evoke adverse events due to systemic T-cell activation. We engineered a bispecific, mAb targeting CD28 homolog (CD28H), a newly identified B7 family receptor that is constitutively expressed on T and natural killer (NK) cells, with a PD-L1 antibody to potentiate tumor-specific immune responses. The bispecific antibody led to T-cell costimulation, induced NK-cell cytotoxicity of PD-L1-expressing tumor cells, and activated tissue-resident memory CD8+ T cells. Mechanistically, the CD28H agonistic arm of the bispecific antibody reduced PD-L1/PD-1-induced SHP2 phosphorylation while simultaneously augmenting T-cell receptor signaling by activating the MAPK and AKT pathways. This bispecific approach could be used to target multiple immune cells, including CD8+ T cells, tissue-resident memory T cells, and NK cells, in a tumor-specific manner that may lead to induction of durable, therapeutic antitumor responses.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos Biespecíficos/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Inmunoterapia , Células Asesinas Naturales , Activación de Linfocitos , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
6.
Cancer Discov ; 11(5): 1100-1117, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33419761

RESUMEN

The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1- T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. SIGNIFICANCE: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma de Células Claras/tratamiento farmacológico , Antígeno CTLA-4/metabolismo , Humanos , Inmunoterapia , Neoplasias Renales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Linfocitos T/inmunología
7.
J Mol Signal ; 12: 3, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-30210579

RESUMEN

Myocyte Stress Protein 1 (MS1) is a muscle-specific, stress-responsive, regulator of gene expression. It was originally identified in embryonic mouse heart which showed increased expression in a rat model of left ventricular hypertrophy. To determine if MS1 was responsive to other stresses relevant to cardiac myocyte function, we tested if it could be induced by the metabolic stresses associated with ischaemia/reperfusion injury in cardiac myocytes. We found that metabolic stress increased MS1 expression, both at the mRNA and protein level, concurrent with activation of the c-Jun N-terminal Kinase (JNK) signalling pathway. MS1 induction by metabolic stress was blocked by both the transcription inhibitor actinomycin D and a JNK inhibitor, suggesting that activation of the JNK pathway during metabolic stress in cardiac myocytes leads to transcriptional induction of MS1. MS1 was also found to be an efficient JNK substrate in vitro, with a major JNK phosphorylation site identified at Thr-62. In addition, MS1 was found to co-precipitate with JNK, and inspection of the amino acid sequence upstream of the phosphorylation site, at Thr-62, revealed a putative Mitogen-Activated Protein Kinase (MAPK) binding site. Taken together, these data identify MS1 as a likely transcriptional and post-translational target for the JNK pathway in cardiac myocytes subjected to metabolic stress.

8.
ACS Appl Mater Interfaces ; 8(1): 974-82, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26698297

RESUMEN

Prompted by the rapidly developing field of wearable electronics, research into biocompatible substrates and coatings is intensifying. Acrylate-based hydrogel polymers have gained widespread use as biocompatible articles in applications such as contact and intraocular lenses. Surface treatments and/or coatings present one strategy to further enhance the performance of these hydrogels or even realize novel functionality. In this study, the conductive polymer poly(3,4-ethylenedioxythiophene) (PEDOT) is deposited from the vapor phase onto hydrated hydrogel substrates and blended with biocompatibilizing coconstituents incorporating polyethylene glycol (PEG) and polydimethyl siloxane (PDMS) moieties. Plasma pretreatment of the dehydrated hydrogel substrate modifies its surface topography and chemical composition to facilitate the attachment of conductive PEDOT-based surface layers. Manipulating the vapor phase polymerization process and constituent composition, the PEDOT-based coating is engineered to be both hydrophilic (i.e. to promote biocompatibility) and highly conductive. The fabrication of this conductively coated hydrogel has implications for the future of wearable electronic devices.


Asunto(s)
Hidrogeles/química , Interacciones Hidrofóbicas e Hidrofílicas , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Dimetilpolisiloxanos/química , Conductividad Eléctrica , Electrodos , Microscopía de Fuerza Atómica , Gases em Plasma/química , Polietilenglicoles/química , Polímeros/química
9.
Cancer Cell ; 24(4): 466-80, 2013 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-24075834

RESUMEN

Aberrant expression of embryonic epithelial-mesenchymal transition-inducing transcription factors (EMT-TFs) in epithelial cells triggers EMT, neoplastic transformation, stemness, and metastatic dissemination. We found that regulation and functions of EMT-TFs are different in malignant melanoma. SNAIL2 and ZEB2 transcription factors are expressed in normal melanocytes and behave as tumor-suppressor proteins by activating an MITF-dependent melanocyte differentiation program. In response to NRAS/BRAF activation, EMT-TF network undergoes a profound reorganization in favor of TWIST1 and ZEB1. This reversible switch cooperates with BRAF in promoting dedifferentiation and neoplastic transformation of melanocytes. We detected EMT-TF reprogramming in late-stage melanoma in association with enhanced phospho-ERK levels. This switch results in E-cadherin loss, enhanced invasion, and constitutes an independent factor of poor prognosis in melanoma patients.


Asunto(s)
Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Animales , Antígenos CD , Cadherinas/metabolismo , Diferenciación Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Melanocitos/citología , Ratones , Ratones Desnudos , Proteínas Nucleares/metabolismo , Fosforilación , Pronóstico , Factores de Transcripción/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
10.
Int J Cancer ; 132(4): 745-54, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22753312

RESUMEN

Embryonic differentiation programs of epithelial-mesenchymal and mesenchymal-epithelial transition (EMT and MET) represent a mechanistic basis for epithelial cell plasticity implicated in cancer. Transcription factors of the ZEB protein family (ZEB1 and ZEB2) and several microRNA species (predominantly miR-200 family members) form a double negative feedback loop, which controls EMT and MET programs in both development and tumorigenesis. In this article, we review crosstalk between the ZEB/miR-200 axis and several signal transduction pathways activated at different stages of tumor development. The close association of ZEB proteins with these pathways is indirect evidence for the involvement of a ZEB/miR-200 loop in tumor initiation, progression and spread. Additionally, the configuration of signaling pathways involving ZEB/miR-200 loop suggests that ZEB1 and ZEB2 may have different, possibly even opposing, roles in some forms of human cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
11.
PLoS Genet ; 8(5): e1002723, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654675

RESUMEN

The epithelial-mesenchymal transition (EMT) is an embryonic transdifferentiation process consisting of conversion of polarized epithelial cells to motile mesenchymal ones. EMT-inducing transcription factors are aberrantly expressed in multiple tumor types and are known to favor the metastatic dissemination process. Supporting oncogenic activity within primary lesions, the TWIST and ZEB proteins can prevent cells from undergoing oncogene-induced senescence and apoptosis by abolishing both p53- and RB-dependent pathways. Here we show that they also downregulate PP2A phosphatase activity and efficiently cooperate with an oncogenic version of H-RAS in malignant transformation of human mammary epithelial cells. Thus, by down-regulating crucial tumor suppressor functions, EMT inducers make cells particularly prone to malignant conversion. Importantly, by analyzing transformed cells generated in vitro and by characterizing novel transgenic mouse models, we further demonstrate that cooperation between an EMT inducer and an active form of RAS is sufficient to trigger transformation of mammary epithelial cells into malignant cells exhibiting all the characteristic features of claudin-low tumors, including low expression of tight and adherens junction genes, EMT traits, and stem cell-like characteristics. Claudin-low tumors are believed to be the most primitive breast malignancies, having arisen through transformation of an early epithelial precursor with inherent stemness properties and metaplastic features. Challenging this prevailing view, we propose that these aggressive tumors arise from cells committed to luminal differentiation, through a process driven by EMT inducers and combining malignant transformation and transdifferentiation.


Asunto(s)
Neoplasias de la Mama , Transformación Celular Neoplásica , Claudinas , Transición Epitelial-Mesenquimal , Glándulas Mamarias Humanas/metabolismo , Proteína Fosfatasa 2 , Proteína 1 Relacionada con Twist/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Claudinas/genética , Claudinas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genes ras , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/citología , Ratones , Ratones Transgénicos , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Proteína de Retinoblastoma/metabolismo , Telomerasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
J Recept Signal Transduct Res ; 31(6): 408-15, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22070384

RESUMEN

ASK1 is a cellular stress-responsive MAPKKK which activates the JNK and p38 MAPK pathways that play a key role in the response of cardiac myocytes to redox stress following ischemia/reperfusion. ASK1 becomes incorporated into high-molecular weight complexes upon activation but this has not been investigated in cardiac myocytes. Here we examine the distribution of ASK1 in neonatal rat cardiomyocytes undergoing simulated ischemia and reperfusion. Simulated ischemia or redox stress in neonatal cardiac myocytes causes the translocation of ASK1 to distinct punctate cytoplasmic structures that are insoluble in Triton X-100. The translocation event is not dependent on ASK1 kinase activity, occurs subsequent to activation and is reversible upon removal of the cell stress. The structures to which ASK1 translocates in cardiac myocytes do not appear to correspond to the previously described ASK1 signalosome reported in other cell types.


Asunto(s)
Citoplasma/enzimología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Animales , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa Quinasa 5/química , Octoxinol/química , Estrés Oxidativo , Fosforilación , Transporte de Proteínas , Ratas , Transducción de Señal , Solubilidad , Estrés Fisiológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Cell Sci ; 124(Pt 13): 2200-7, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21652629

RESUMEN

Heterozygous mutations of p63, a key transcription factor in epithelial development, are causative in a variety of human ectodermal dysplasia disorders. Although the mutation spectrum of these disorders displays a striking genotype-phenotype association, the molecular basis for this association is only superficially known. Here, we characterize the transcriptional activity and protein stability of ΔNp63 mutants (that is, mutants of a p63 isoform that lacks the N-terminal transactivation domain) that are found in ectrodactyly-ectodermal dysplasia-cleft syndrome (EEC), ankyloblepharon-ectodermal dysplasia-clefting syndrome (AEC) and nonsyndromic split-hand/split-foot malformation (SHFM). DNA-binding and sterile alpha motif (SAM) domain mutants accumulate in the skin of EEC and AEC syndrome patients, respectively, and show extended half lives in vitro. By contrast, C-terminal mutations found in SHFM patients have half-lives similar to that of the wild-type protein. The increased half-life of EEC and AEC mutant proteins was reverted by overexpression of wild-type ΔNp63. Interestingly, the mutant proteins exhibit normal binding to and degradation by the E3 ubiquitin ligase Itch. Finally, EEC and AEC mutant proteins have reduced transcriptional activity on several skin-specific gene promoters, whereas SHFM mutant proteins are transcriptionally active. Our results, therefore, provide evidence for a regulatory feedback mechanism for p63 that links transcriptional activity to regulation of protein homeostasis by an unknown mechanism. Disruption of this regulatory mechanism might contribute to the pathology of p63-related developmental disorders.


Asunto(s)
Displasia Ectodérmica/genética , Proteínas de la Membrana/metabolismo , Activación Transcripcional/genética , Labio Leporino/genética , Fisura del Paladar/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Anomalías del Ojo/genética , Párpados/anomalías , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Células HEK293 , Semivida , Humanos , Deformidades Congénitas de las Extremidades/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidad Proteica , Receptores de LDL/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
14.
Biochem Biophys Res Commun ; 401(4): 568-73, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20888799

RESUMEN

The formation of a stratified epidermis requires a carefully controlled balance between keratinocyte proliferation and differentiation. Here, we report the reciprocal effect on keratin expression of ΔNp63, pivotal in normal epidermal morphogenesis and maintenance, and Skn-1a/Oct-11, a POU transcription factor that triggers and regulates the differentiation of keratinocytes. The expression of Skn-1a markedly downregulated ΔNp63-driven K14 expression in luciferase reporter assays. The extent of downregulation was comparable to the inhibition of Skn-1a-mediated K10 expression upon expression of ΔNp63. ΔNp63, mutated in the protein-protein interaction domain (SAM domain; mutated in human ectodermal dysplasia syndrome), was significantly less effecting in downregulating K10, raising the possibility of a direct interaction among Skn-1a and ΔNp63. Immunolocalization in human skin biopsies revealed that the expression of the two transcription factors is partially overlapping. Co-immunoprecipitation experiments did not, however, demonstrate a direct interaction between ΔNp63 and Skn-1a, suggesting that the antagonistic effects of Skn-1a and p63 on keratin promoter transactivation is probably through competition for overlapping binding sites on target gene promoter or through an indirect interaction.


Asunto(s)
Epidermis/fisiología , Queratinocitos/fisiología , Queratinas/genética , Factores de Transcripción de Octámeros/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Proteínas Supresoras de Tumor/metabolismo , Diferenciación Celular/genética , Línea Celular , Células Epidérmicas , Humanos , Inmunoprecipitación , Queratina-10/antagonistas & inhibidores , Queratina-10/genética , Queratina-14/antagonistas & inhibidores , Queratina-14/genética , Queratinocitos/citología , Queratinas/antagonistas & inhibidores , Factores de Transcripción de Octámeros/genética , Regiones Promotoras Genéticas , Transactivadores/genética , Factores de Transcripción , Proteínas Supresoras de Tumor/genética
15.
Proc Natl Acad Sci U S A ; 107(29): 12877-82, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20615966

RESUMEN

p73 is a p53-related transcription factor with fundamental roles in development and tumor suppression. Transcription from two different promoters on the p73 gene results in generation of transcriptionally active TAp73 isoforms and dominant negative DeltaNp73 isoforms with opposing pro- and anti-apoptotic functions. Therefore, the relative ratio of each isoform is an important determinant of the cell fate. Proteasomal degradation of p73 is mediated by polyubiquitination-dependent and -independent processes both of which appear, thus far, to lack selectivity for the TAp73 and DeltaNp73 isoforms. Here, we describe the characterization of another transcriptional target of TAp73; a ring finger domain ubiquitin ligase p73 Induced RING 2 protein (PIR2). Although PIR2 was initially identified a p53-induced gene (p53RFP), low abundance of PIR2 transcript in mouse embryonic fibroblasts of TAp73 KO mice compared with WT mice and comparison of PIR2 mRNA and protein levels following TAp73 or p53 overexpression substantiate TAp73 isoforms as strong inducers of PIR2. Although PIR2 expression was induced by DNA damage, its expression did not alter apoptotic response or cell cycle profile per se. However, coexpression of PIR2 with TAp73 or DeltaNp73 resulted in an increase of the TA/DeltaNp73 ratio, due to preferential degradation of DeltaNp73. Finally, PIR2 was able to relieve the inhibitory effect of DeltaNp73 on TAp73 induced apoptosis following DNA damage. These results suggest that PIR2, by being induced by TAp73 and degrading DeltaNp73, differentially regulates TAp73/DeltaNp73 stability, and, hence, it may offer a therapeutic approach to enhance the chemosensitivity of tumor cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Dominios RING Finger , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Daño del ADN , Células HCT116 , Humanos , Ratones , Unión Proteica , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteína Tumoral p73 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
16.
Biochim Biophys Acta ; 1802(9): 733-40, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20550965

RESUMEN

p38 MAPK is activated potently during cardiac ischaemia, although the precise mechanism by which it is activated is unclear. We used the isolated perfused rat heart to investigate the signalling pathways activated upstream of p38 during global cardiac ischaemia. Ischaemia strongly activated p38alpha but not the JNK pathway. The MAPKKs, MKK3, MKK4 and MKK6 have previously been identified as potential upstream activators of p38; however, in the ischaemic perfused heart, we saw activation of MKK3 and MKK6 but not MKK4. MKK3 and MKK6 showed different temporal patterns of activity, indicating distinct modes of activation and physiological function. Consistent with a lack of JNK activation, we saw no activation of MKK4 or MKK7 at any time point during ischaemia. A lack of MKK4 activation indicates, at least in the ischaemic heart, that MKK4 is not a physiologically relevant activator of p38. The MAPKKK, ASK1, was strongly activated late during ischaemia, with a similar time course to that of MKK6 and in ischaemic neonatal cardiac myocytes ASK1 expression preferentially activated MKK6 rather than MKK3. These observations suggest that during ischaemia ASK1 is coupled to p38 activation primarily via MKK6. Potent activation of ASK1 during ischaemia without JNK activation shows that during cardiac ischaemia, ASK1 preferentially activates the p38 pathway. These results demonstrate a specificity of responses seldom seen in previous studies and illustrate the benefits of using direct assays in intact tissues responding to physiologically relevant stimuli to unravel the complexities of MAPK signalling.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Isquemia Miocárdica/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática/fisiología , Humanos , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Modelos Biológicos , Isquemia Miocárdica/patología , Ratas , Ratas Sprague-Dawley , Ratas Wistar
17.
Cell Cycle ; 9(5): 886-91, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20160487

RESUMEN

Epithelial mesenchymal transitions (EMT), the generation of motile mesenchymal cells from epithelial sheets, are differentiation programs which take place at several critical steps of embryonic development and in metastatic cancer. Recent data have shown that the transcription factors which are master regulators of EMT also regulate cell cycle progression, apoptosis and senescence. In light of these new observations, the role of these factors in human cancer may be broader than previously anticipated. Here we review recent literature on non-EMT functions of EMT-controlling transcription factors. We will mainly focus on transcription factors belonging to the ZEB family, but some important results obtained by investigators studying other key EMT regulators, Snail and Twist are also discussed.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Apoptosis , Ciclo Celular , Diferenciación Celular , Movimiento Celular , Senescencia Celular , Células Epiteliales/citología , Humanos , Neoplasias/etiología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Wnt/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
18.
Proc Natl Acad Sci U S A ; 106(35): 14884-9, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706487

RESUMEN

The epithelial-mesenchymal transition (EMT) contributes to cancer metastasis. Two ZEB family members, ZEB1 and ZEB2(SIP1), inhibit transcription of the E-cadherin gene and induce EMT in vitro. However, their relevance to human cancer is insufficiently studied. Here, we performed a comparative study of SIP1 and ZEB1 proteins in cancer cell lines and in one form of human malignancy, carcinoma of the bladder. Whereas ZEB1 protein was expressed in all E-cadherin-negative carcinoma cell lines, being in part responsible for the high motility of bladder cancer cells, SIP1 was hardly ever detectable in carcinoma cells in culture. However, SIP1 represented an independent factor of poor prognosis (P = 0.005) in a series of bladder cancer specimens obtained from patients treated with radiotherapy. In contrast, ZEB1 was rarely expressed in tumor tissues; and E-cadherin status did not correlate with the patients' survival. SIP1 protected cells from UV- and cisplatin-induced apoptosis in vitro but had no effect on the level of DNA damage. The anti-apoptotic effect of SIP1 was independent of either cell cycle arrest or loss of cell-cell adhesion and was associated with reduced phosphorylation of ATM/ATR targets in UV-treated cells. The prognostic value of SIP1 and its role in DNA damage response establish a link between genetic instability and metastasis and suggest a potential importance for this protein as a therapeutic target. In addition, we conclude that the nature of an EMT pathway rather than the deregulation of E-cadherin per se is critical for the progression of the disease and patients' survival.


Asunto(s)
Apoptosis , Daño del ADN , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Invasividad Neoplásica , Fenotipo , Pronóstico , Proteínas Represoras/genética , Tasa de Supervivencia , Factores de Transcripción/metabolismo , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/radioterapia , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
19.
Biochem J ; 402(1): 187-96, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17123353

RESUMEN

PP1 (protein phosphatase 1) is a ubiquitously expressed serine/threonine-specific protein phosphatase whose activity towards different substrates appears to be mediated via binding to specific proteins that play critical regulatory and targeting roles. In the present paper we report the cloning and characterization of a new protein, termed SARP (several ankyrin repeat protein), which is shown to interact with all isoforms of PP1 by a variety of techniques. A region encompassing a consensus PP1-binding motif in SARP (K354VHF357) modulates endogenous SARP-PP1 activity in mammalian cells. This SARP-PP1 interaction motif lies partially within the first ankyrin repeat in contrast with other proteins [53BP2 (p53 binding protein 2), MYPT1/M(110)/MBS (myosin binding protein of PP1) and TIMAP (transforming growth factor beta inhibited, membrane-associated protein)], where a PP1-binding motif precedes the ankyrin repeats. Alternative mRNA splicing produces several isoforms of SARP from a single human gene at locus 11q14. SARP1 and/or SARP2 (92-95 kDa) are ubiquitously expressed in all tissues with high levels in testis and sperm, where they are shown to interact with both PP1gamma1 and PP1gamma2. SARP3 (65 kDa) is most abundant in brain where SARP isoforms interact with both PP1alpha and PP1gamma1. SARP is highly abundant in the nucleus of mammalian cells, consistent with the putative nuclear localization signal at the N-terminus. The presence of a leucine zipper near the C-terminus of SARP1 and SARP2, and the binding of mammalian DNA to SARP2, suggests that SARP1 and SARP2 may be transcription factors or DNA-associated proteins that modulate gene expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Repetición de Anquirina , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Clonación Molecular , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas del Ojo/química , Proteínas del Ojo/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Leucina Zippers , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Fosfatasa 1 , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
20.
J Biol Chem ; 280(19): 18717-27, 2005 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15772076

RESUMEN

Amino acids positively regulate signaling through the mammalian target of rapamycin (mTOR). Recent work demonstrated the importance of the tuberous sclerosis protein TSC2 for regulation of mTOR by insulin. TSC2 contains a GTPase-activator domain that promotes hydrolysis of GTP bound to Rheb, which positively regulates mTOR signaling. Some studies have suggested that TSC2 also mediates the control of mTOR by amino acids. In cells lacking TSC2, amino acid withdrawal still results in dephosphorylation of S6K1, ribosomal protein S6, the eukaryotic initiation factor 4E-binding protein, and elongation factor-2 kinase. The effects of amino acid withdrawal are diminished by inhibiting protein synthesis or adding back amino acids. These studies demonstrate that amino acid signaling to mTOR occurs independently of TSC2 and involves additional unidentified inputs. Although TSC2 is not required for amino acid control of mTOR, amino acid withdrawal does decrease the proportion of Rheb in the active GTP-bound state. Here we also show that Rheb and mTOR form stable complexes, which are not, however, disrupted by amino acid withdrawal. Mutants of Rheb that cannot bind GTP or GDP can interact with mTOR complexes. We also show that the effects of hydrogen peroxide and sorbitol, cell stresses that impair mTOR signaling, are independent of TSC2. Finally, we show that the ability of energy depletion (which impairs mTOR signaling in TSC2+/+ cells) to increase the phosphorylation of eukaryotic elongation factor 2 is also independent of TSC2. This likely involves the phosphorylation of the elongation factor-2 kinase by the AMP-activated protein kinase.


Asunto(s)
Aminoácidos/metabolismo , Proteínas de Unión al GTP Monoméricas/fisiología , Neuropéptidos/fisiología , Proteínas Quinasas/metabolismo , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas Quinasas Activadas por AMP , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfato/química , Aminoácidos/química , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Portadoras/química , Proteínas de Ciclo Celular , Línea Celular , Células Cultivadas , Cicloheximida/farmacología , Relación Dosis-Respuesta a Droga , Quinasa del Factor 2 de Elongación , Factores Eucarióticos de Iniciación , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Glucosa/química , Guanina/química , Guanosina Difosfato/química , Guanosina Trifosfato/química , Humanos , Peróxido de Hidrógeno/farmacología , Hidrólisis , Immunoblotting , Inmunoprecipitación , Ratones , Modelos Biológicos , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multienzimáticos/metabolismo , Mutación , Neuropéptidos/química , Neuropéptidos/metabolismo , Fosfoproteínas/química , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Inhibidores de la Síntesis de la Proteína/farmacología , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteínas Represoras/metabolismo , Proteína S6 Ribosómica/metabolismo , Transducción de Señal , Sorbitol/farmacología , Serina-Treonina Quinasas TOR , Factores de Tiempo , Transgenes , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA