Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Photonics ; 11(4): 1592-1603, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38645993

RESUMO

Integrated single-molecule force-fluorescence spectroscopy setups allow for simultaneous fluorescence imaging and mechanical force manipulation and measurements on individual molecules, providing comprehensive dynamic and spatiotemporal information. Dual-beam optical tweezers (OT) combined with a confocal scanning microscope form a force-fluorescence spectroscopy apparatus broadly used to investigate various biological processes, in particular, protein:DNA interactions. Such experiments typically involve imaging of fluorescently labeled proteins bound to DNA and force spectroscopy measurements of trapped individual DNA molecules. Here, we present a versatile state-of-the-art toolbox including the preparation of protein:DNA complex samples, design of a microfluidic flow cell incorporated with OT, automation of OT-confocal scanning measurements, and the development and implementation of a streamlined data analysis package for force and fluorescence spectroscopy data processing. Its components can be adapted to any commercialized or home-built dual-beam OT setup equipped with a confocal scanning microscope, which will facilitate single-molecule force-fluorescence spectroscopy studies on a large variety of biological systems.

2.
Nat Commun ; 14(1): 6735, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37872142

RESUMO

Chromatin replication involves the assembly and activity of the replisome within the nucleosomal landscape. At the core of the replisome is the Mcm2-7 complex (MCM), which is loaded onto DNA after binding to the Origin Recognition Complex (ORC). In yeast, ORC is a dynamic protein that diffuses rapidly along DNA, unless halted by origin recognition sequences. However, less is known about the dynamics of ORC proteins in the presence of nucleosomes and attendant consequences for MCM loading. To address this, we harnessed an in vitro single-molecule approach to interrogate a chromatinized origin of replication. We find that ORC binds the origin of replication with similar efficiency independently of whether the origin is chromatinized, despite ORC mobility being reduced by the presence of nucleosomes. Recruitment of MCM also proceeds efficiently on a chromatinized origin, but subsequent movement of MCM away from the origin is severely constrained. These findings suggest that chromatinized origins in yeast are essential for the local retention of MCM, which may facilitate subsequent assembly of the replisome.


Assuntos
Complexo de Reconhecimento de Origem , Proteínas de Saccharomyces cerevisiae , Complexo de Reconhecimento de Origem/genética , Complexo de Reconhecimento de Origem/metabolismo , Nucleossomos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Replicação do DNA , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Origem de Replicação
3.
Nat Commun ; 14(1): 2082, 2023 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-37059705

RESUMO

The eukaryotic replicative helicase CMG centrally orchestrates the replisome and leads the way at the front of replication forks. Understanding the motion of CMG on the DNA is therefore key to our understanding of DNA replication. In vivo, CMG is assembled and activated through a cell-cycle-regulated mechanism involving 36 polypeptides that has been reconstituted from purified proteins in ensemble biochemical studies. Conversely, single-molecule studies of CMG motion have thus far relied on pre-formed CMG assembled through an unknown mechanism upon overexpression of individual constituents. Here, we report the activation of CMG fully reconstituted from purified yeast proteins and the quantification of its motion at the single-molecule level. We observe that CMG can move on DNA in two ways: by unidirectional translocation and by diffusion. We demonstrate that CMG preferentially exhibits unidirectional translocation in the presence of ATP, whereas it preferentially exhibits diffusive motion in the absence of ATP. We also demonstrate that nucleotide binding halts diffusive CMG independently of DNA melting. Taken together, our findings support a mechanism by which nucleotide binding allows newly assembled CMG to engage with the DNA within its central channel, halting its diffusion and facilitating the initial DNA melting required to initiate DNA replication.


Assuntos
Eucariotos , Nucleotídeos , Eucariotos/metabolismo , Replicação do DNA , DNA Helicases/metabolismo , DNA/metabolismo , Trifosfato de Adenosina/metabolismo
4.
Nucleic Acids Res ; 51(8): 3770-3792, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36942484

RESUMO

During every cell cycle, both the genome and the associated chromatin must be accurately replicated. Chromatin Assembly Factor-1 (CAF-1) is a key regulator of chromatin replication, but how CAF-1 functions in relation to the DNA replication machinery is unknown. Here, we reveal that this crosstalk differs between the leading and lagging strand at replication forks. Using biochemical reconstitutions, we show that DNA and histones promote CAF-1 recruitment to its binding partner PCNA and reveal that two CAF-1 complexes are required for efficient nucleosome assembly under these conditions. Remarkably, in the context of the replisome, CAF-1 competes with the leading strand DNA polymerase epsilon (Polϵ) for PCNA binding. However, CAF-1 does not affect the activity of the lagging strand DNA polymerase Delta (Polδ). Yet, in cells, CAF-1 deposits newly synthesized histones equally on both daughter strands. Thus, on the leading strand, chromatin assembly by CAF-1 cannot occur simultaneously to DNA synthesis, while on the lagging strand these processes may be coupled. We propose that these differences may facilitate distinct parental histone recycling mechanisms and accommodate the inherent asymmetry of DNA replication.


Assuntos
Cromatina , Histonas , Histonas/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fator 1 de Modelagem da Cromatina/genética , Fator 1 de Modelagem da Cromatina/metabolismo , Cromatina/genética , Replicação do DNA , DNA/genética
5.
Cell Rep ; 41(3): 111491, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36261017

RESUMO

Ring-shaped structural maintenance of chromosomes (SMC) complexes like condensin and cohesin extrude loops of DNA. It remains, however, unclear how they can extrude DNA loops in chromatin that is bound with proteins. Here, we use in vitro single-molecule visualization to show that nucleosomes, RNA polymerase, and dCas9 pose virtually no barrier to loop extrusion by yeast condensin. We find that even DNA-bound nanoparticles as large as 200 nm, much bigger than the SMC ring size, also translocate into DNA loops during extrusion by condensin and cohesin. This even occurs for a single-chain version of cohesin in which the ring-forming subunits are covalently linked and cannot open to entrap DNA. The data show that SMC-driven loop extrusion has surprisingly little difficulty in accommodating large roadblocks into the loop. The findings also show that the extruded DNA does not pass through the SMC ring (pseudo)topologically, hence pointing to a nontopological mechanism for DNA loop extrusion.


Assuntos
Nanopartículas , Nucleossomos , Proteínas de Ciclo Celular , Cromatina , Saccharomyces cerevisiae
6.
STAR Protoc ; 3(3): 101606, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-35959497

RESUMO

Multiplexed single-molecule magnetic tweezers (MT) have recently been employed to probe the RNA synthesis dynamics of RNA-dependent RNA polymerases (RdRp). Here, we present a protocol for simultaneously probing the RNA synthesis dynamics of hundreds of single polymerases with MT. We describe the preparation of a dsRNA construct for probing single RdRp kinetics. We then detail the measurement of RdRp RNA synthesis kinetics using MT. The protocol is suitable for high-throughput probing of RdRp-targeting antiviral compounds for mechanistic function and efficacy. For complete details on the use and execution of this protocol, please refer to Janissen et al. (2021).


Assuntos
Antivirais , RNA Polimerase Dependente de RNA , Cinética , Fenômenos Magnéticos , RNA de Cadeia Dupla
7.
Nat Commun ; 12(1): 1908, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33772005

RESUMO

DNA replication in eukaryotes initiates at many origins distributed across each chromosome. Origins are bound by the origin recognition complex (ORC), which, with Cdc6 and Cdt1, recruits and loads the Mcm2-7 (MCM) helicase as an inactive double hexamer during G1 phase. The replisome assembles at the activated helicase in S phase. Although the outline of replisome assembly is understood, little is known about the dynamics of individual proteins on DNA and how these contribute to proper complex formation. Here we show, using single-molecule optical trapping and confocal microscopy, that yeast ORC is a mobile protein that diffuses rapidly along DNA. Origin recognition halts this search process. Recruitment of MCM molecules in an ORC- and Cdc6-dependent fashion results in slow-moving ORC-MCM intermediates and MCMs that rapidly scan the DNA. Following ATP hydrolysis, salt-stable loading of MCM single and double hexamers was seen, both of which exhibit salt-dependent mobility. Our results demonstrate that effective helicase loading relies on an interplay between protein diffusion and origin recognition, and suggest that MCM is stably loaded onto DNA in multiple forms.


Assuntos
Proteínas de Ciclo Celular/genética , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Manutenção de Minicromossomo/genética , Complexo de Reconhecimento de Origem/genética , Origem de Replicação/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Algoritmos , Sítios de Ligação/genética , Proteínas de Ciclo Celular/metabolismo , DNA Fúngico/genética , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Modelos Genéticos , Complexo de Reconhecimento de Origem/metabolismo , Ligação Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
Cell Rep ; 21(4): 1063-1076, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-29069588

RESUMO

RNA viruses pose a threat to public health that is exacerbated by the dearth of antiviral therapeutics. The RNA-dependent RNA polymerase (RdRp) holds promise as a broad-spectrum, therapeutic target because of the conserved nature of the nucleotide-substrate-binding and catalytic sites. Conventional, quantitative, kinetic analysis of antiviral ribonucleotides monitors one or a few incorporation events. Here, we use a high-throughput magnetic tweezers platform to monitor the elongation dynamics of a prototypical RdRp over thousands of nucleotide-addition cycles in the absence and presence of a suite of nucleotide analog inhibitors. We observe multiple RdRp-RNA elongation complexes; only a subset of which are competent for analog utilization. Incorporation of a pyrazine-carboxamide nucleotide analog, T-1106, leads to RdRp backtracking. This analysis reveals a mechanism of action for this antiviral ribonucleotide that is corroborated by cellular studies. We propose that induced backtracking represents a distinct mechanistic class of antiviral ribonucleotides.


Assuntos
Antivirais/farmacologia , Nucleosídeos/farmacologia , Pirazinas/farmacologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Antivirais/química , Células HeLa , Humanos , Imageamento Tridimensional , Campos Magnéticos , Nucleosídeos/química , Pirazinas/química , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/química , Ribonucleotídeos/química , Ribonucleotídeos/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química
9.
Methods ; 105: 90-8, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27038745

RESUMO

Recent advances in high-throughput single-molecule magnetic tweezers have paved the way for obtaining information on individual molecules as well as ensemble-averaged behavior in a single assay. Here we describe how to design robust high-throughput magnetic tweezers assays that specifically require application of high forces (>20pN) for prolonged periods of time (>1000s). We elaborate on the strengths and limitations of the typical construct types that can be used and provide a step-by-step guide towards a high tether yield assay based on two examples. Firstly, we discuss a DNA hairpin assay where force-induced strand separation triggers a tight interaction between DNA-binding protein Tus and its binding site Ter, where forces up to 90pN for hundreds of seconds were required to dissociate Tus from Ter. Secondly, we show how the LTag helicase of Simian virus 40 unwinds dsDNA, where a load of 36pN optimizes the assay readout. The approaches detailed here provide guidelines for the high-throughput, quantitative study of a wide range of DNA-protein interactions.


Assuntos
DNA Helicases/química , Proteínas de Ligação a DNA/química , Ensaios de Triagem em Larga Escala/métodos , Imagem Individual de Molécula/métodos , DNA/química , DNA Helicases/isolamento & purificação , Proteínas de Ligação a DNA/genética , Pinças Ópticas , Vírus 40 dos Símios/enzimologia
10.
J Biol Eng ; 9: 11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26140052

RESUMO

BACKGROUND: Chromosome engineering encompasses a collection of homologous recombination-based techniques that are employed to modify the genome of a model organism in a controlled fashion. Such techniques are widely used in both fundamental and industrial research to introduce multiple insertions in the same Escherichia coli strain. To date, λ-Red recombination (also known as recombineering) and P1 phage transduction are the most successfully implemented chromosome engineering techniques in E. coli. However, due to errors that can occur during the strain creation process, reliable validation methods are essential upon alteration of a strain's chromosome. RESULTS AND DISCUSSION: Polymerase chain reaction (PCR)-based methods and DNA sequence analysis are rapid and powerful methods to verify successful integration of DNA sequences into a chromosome. Even though these verification methods are necessary, they may not be sufficient in detecting all errors, imposing the requirement of additional validation methods. For example, as extraneous insertions may occur during recombineering, we highlight the use of Southern blotting to detect their presence. These unwanted mutations can be removed via transducing the region of interest into the wild type chromosome using P1 phages. However, in doing so one must verify that both the P1 lysate and the strains utilized are free from contamination with temperate phages, as these can lysogenize inside a cell as a large plasmid. Thus, we illustrate various methods to probe for temperate phage contamination, including cross-streak agar and Evans Blue-Uranine (EBU) plate assays, whereby the latter is a newly reported technique for this purpose in E. coli. Lastly, we discuss methodologies for detecting defects in cell growth and shape characteristics, which should be employed as an additional check. CONCLUSION: The simple, yet crucial validation techniques discussed here can be used to reliably verify any chromosomally engineered E. coli strains for errors such as non-specific insertions in the chromosome, temperate phage contamination, and defects in growth and cell shape. While techniques such as PCR and DNA sequence verification should standardly be performed, we illustrate the necessity of performing these additional assays. The discussed techniques are highly generic and can be easily applied to any type of chromosome engineering.

11.
Nat Chem Biol ; 11(8): 579-85, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26147356

RESUMO

The bidirectional replication of a circular chromosome by many bacteria necessitates proper termination to avoid the head-on collision of the opposing replisomes. In Escherichia coli, replisome progression beyond the termination site is prevented by Tus proteins bound to asymmetric Ter sites. Structural evidence indicates that strand separation on the blocking (nonpermissive) side of Tus-Ter triggers roadblock formation, but biochemical evidence also suggests roles for protein-protein interactions. Here DNA unzipping experiments demonstrate that nonpermissively oriented Tus-Ter forms a tight lock in the absence of replicative proteins, whereas permissively oriented Tus-Ter allows nearly unhindered strand separation. Quantifying the lock strength reveals the existence of several intermediate lock states that are impacted by mutations in the lock domain but not by mutations in the DNA-binding domain. Lock formation is highly specific and exceeds reported in vivo efficiencies. We postulate that protein-protein interactions may actually hinder, rather than promote, proper lock formation.


Assuntos
Replicação do DNA , DNA Bacteriano/metabolismo , DNA Circular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Sequência de Bases , Sítios de Ligação , Cromossomos Bacterianos/química , Cromossomos Bacterianos/metabolismo , DNA Bacteriano/química , DNA Circular/química , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
12.
Breast Cancer Res ; 17: 28, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25849225

RESUMO

INTRODUCTION: Increased expression of αv integrins is frequently associated with tumor cell adhesion, migration, invasion and metastasis, and correlates with poor prognosis in breast cancer. However, the mechanism by which αv integrins can enhance breast cancer progression is still largely unclear. The effects of therapeutic targeting of αv integrins in breast cancer also have yet to be investigated. METHODS: We knocked down αv integrin in MDA-MB-231 and MCF10A-M4 breast cancer cells, or treated these cells with the αv antagonist GLPG0187. The effects of αv integrin depletion on mesenchymal markers, transforming growth factor-ß (TGF-ß)/Smad signaling and TGF-ß-induced target gene expression were analyzed in MDA-MB-231 cells by RNA analysis or Western blotting. The function of αv integrin on breast cancer cell migration was investigated by transwell assay in vitro, and its effect on breast cancer progression was assessed by both zebrafish and mouse xenografts in vivo. In the mouse model, GLPG0187 was administered separately, or in combination with the standard-of-care anti-resorptive agent zoledronate and the chemotherapeutic drug paclitaxel, to study the effects of combinational treatments on breast cancer metastasis. RESULTS: Genetic interference and pharmacological targeting of αv integrin with GLPG0187 in different breast cancer cell lines inhibited invasion and metastasis in the zebrafish or mouse xenograft model. Depletion of αv integrin in MDA-MB-231 cells inhibited the expression of mesenchymal markers and the TGF-ß/Smad response. TGF-ß induced αv integrin mRNA expression and αv integrin was required for TGF-ß-induced breast cancer cell migration. Moreover, treatment of MDA-MB-231 cells with non-peptide RGD antagonist GLPG0187 decreased TGF-ß signaling. In the mouse xenografts GLPG0187 inhibited the progression of bone metastasis. Maximum efficacy of inhibition of bone metastasis was achieved when GLPG0187 was combined with the standard-of-care metastatic breast cancer treatments. CONCLUSION: These findings show that αv integrin is required for efficient TGF-ß/Smad signaling and TGF-ß-induced breast cancer cell migration, and for maintaining a mesenchymal phenotype of the breast cancer cells. Our results also provide evidence that targeting αv integrin could be an effective therapeutic approach for treatment of breast cancer tumors and/or metastases that overexpress αv integrin.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Deleção de Genes , Integrina alfaV/genética , Animais , Animais Geneticamente Modificados , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Integrina alfaV/metabolismo , Camundongos , Metástase Neoplásica , Fator de Crescimento Transformador beta/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
13.
Nucleic Acids Res ; 42(18): e137, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25140010

RESUMO

Magnetic tweezers are a powerful single-molecule technique that allows real-time quantitative investigation of biomolecular processes under applied force. High pulling forces exceeding tens of picoNewtons may be required, e.g. to probe the force range of proteins that actively transcribe or package the genome. Frequently, however, the application of such forces decreases the sample lifetime, hindering data acquisition. To provide experimentally viable sample lifetimes in the face of high pulling forces, we have designed a novel anchoring strategy for DNA in magnetic tweezers. Our approach, which exploits covalent functionalization based on heterobifunctional poly(ethylene glycol) crosslinkers, allows us to strongly tether DNA while simultaneously suppressing undesirable non-specific adhesion. A complete force and lifetime characterization of these covalently anchored DNA-tethers demonstrates that, compared to more commonly employed anchoring strategies, they withstand 3-fold higher pulling forces (up to 150 pN) and exhibit up to 200-fold higher lifetimes (exceeding 24 h at a constant force of 150 pN). This advance makes it possible to apply the full range of biologically relevant force scales to biomolecular processes, and its straightforward implementation should extend its reach to a multitude of applications in the field of single-molecule force spectroscopy.


Assuntos
DNA/química , Fenômenos Biomecânicos , Imãs , Polietilenoglicóis/química , Análise Espectral
14.
Biochem Biophys Res Commun ; 435(1): 58-63, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23618854

RESUMO

TGF-ß plays a dual role in cancer; in early stages it inhibits tumor growth, whereas later it promotes invasion and metastasis. TGF-ß is thought to be pro-invasive by inducing epithelial-to-mesenchymal transition (EMT) via induction of transcriptional repressors, including Slug and Snail. In this study, we investigated the role of Snail and Slug in TGF-ß-induced invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model. Ectopic expression of Slug or Snail promoted invasion of single, rounded amoeboid cells in vitro. In an embryonic zebrafish xenograft model, forced expression of Slug and Snail promoted single cell invasion and metastasis. Slug and Snail are sufficient for the induction of single-cell invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model.


Assuntos
Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Animais , Western Blotting , Linhagem Celular , Movimento Celular/genética , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Transição Epitelial-Mesenquimal/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Peixe-Zebra/embriologia , Peixe-Zebra/genética
15.
Biochim Biophys Acta ; 1823(8): 1233-41, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22613765

RESUMO

ß- and γ-secretase cleave the amyloid precursor protein (APP) to release the amyloidogenic ß-amyloid peptides (Aß) and the APP intracellular domain (AICD). Aß has been widely believed to initiate pathogenic cascades culminating in Alzheimer's disease (AD). However, the physiological functions of the AICD remain elusive. In this study, we found the AICD to strongly inhibit Wnt-induced transcriptional reporter activity, and to counteract Wnt-induced c-Myc expression. Loss of the AICD resulted in an increased responsiveness to Wnt/ß-catenin-mediated transcription. Mechanically, the AICD was found to interact with glycogen synthase kinase 3 beta (GSK3ß) and promote its kinase activity. The subsequent AICD-strengthened Axin-GSK3ß complex potentiates ß-catenin poly-ubiquitination. Functional studies in N(2)a mouse neuroblastoma cells, rat pheochromocytoma PC12 cells and primary neurons showed that the AICD facilitated neurite outgrowth. And AICD antagonised Wnt3a-suppressed growth arrest and neurite outgrowth in N2a and PC12 cells. Taken together, our results identify the AICD as a novel inhibitory factor of the canonical Wnt signalling pathway and suggest its regulatory role in neuronal cell proliferation and differentiation.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Neuritos/fisiologia , Via de Sinalização Wnt , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Proliferação de Células , Expressão Gênica , Regulação da Expressão Gênica , Genes Reporter , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Fator de Crescimento Neural/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Cultura Primária de Células , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
16.
Cell Oncol (Dordr) ; 35(1): 19-28, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21935711

RESUMO

BACKGROUND: The transforming growth factor (TGF)-ß superfamily comprises cytokines such as TGF-ß and Bone Morphogenetic Proteins (BMPs), which have a critical role in a multitude of biological processes. In breast cancer, high levels of TGF-ß are associated with poor outcome, whereas inhibition of TGF-ß-signaling reduces metastasis. In contrast, BMP-7 inhibits bone metastasis of breast cancer cells. METHODS: In this study, we investigated the effect of BMP-7 on TGF-ß-induced invasion in a 3 dimensional invasion assay. RESULTS: BMP-7 inhibited TGF-ß-induced invasion of the metastatic breast cancer cell line MCF10CA1a, but not of its premalignant precursor MCF10AT in a spheroid invasion model. The inhibitory effect appears to be specific for BMP-7, as its closest homolog, BMP-6, did not alter the invasion of MCF10CA1a spheroids. To elucidate the mechanism by which BMP-7 inhibits TGF-ß-induced invasion, we analyzed invasion-related genes. BMP-7 inhibited TGF-ß-induced expression of integrin α(v)ß(3) in the spheroids. Moreover, targeting of integrins by a chemical inhibitor or knockdown of integrin ß(3) negatively affected TGF-ß-induced invasion. On the other hand, overexpression of integrin ß(3) counteracted the inhibitory effect of BMP7 on TGF-ß-induced invasion. CONCLUSION: Thus, BMP-7 may exert anti-invasive actions by inhibiting TGF-ß-induced expression of integrin ß(3).


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Crescimento Transformador beta/farmacologia , Proteína Morfogenética Óssea 6/farmacologia , Proteína Morfogenética Óssea 7/antagonistas & inibidores , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Proteínas de Transporte/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Integrina alfa5/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Transdução de Sinais/genética , Proteínas Smad/metabolismo
17.
J Vis Exp ; (57)2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22126971

RESUMO

TGF-ß has opposing roles in breast cancer progression by acting as a tumor suppressor in the initial phase, but stimulating invasion and metastasis at later stage(1,2). Moreover, TGF-ß is frequently overexpressed in breast cancer and its expression correlates with poor prognosis and metastasis (3,4). The mechanisms by which TGF-ß induces invasion are not well understood. TGF-ß elicits its cellular responses via TGF-ß type II (TßRII) and type I (TßRI) receptors. Upon TGF-ß-induced heteromeric complex formation, TßRII phosphorylates the TßRI. The activated TßRI initiates its intracellular canonical signaling pathway by phosphorylating receptor Smads (R-Smads), i.e. Smad2 and Smad3. These activated R-Smads form heteromeric complexes with Smad4, which accumulate in the nucleus and regulate the transcription of target genes(5). In addition to the previously described Smad pathway, receptor activation results in activation of several other non-Smad signaling pathways, for example Mitogen Activated Protein Kinase (MAPK) pathways(6). To study the role of TGF-ß in different stages of breast cancer, we made use of the MCF10A cell system. This system consists of spontaneously immortalized MCF10A1 (M1) breast epithelial cells(7), the H-RAS transformed M1-derivative MCF10AneoT (M2), which produces premalignant lesions in mice(8), and the M2-derivative MCF10CA1a (M4), which was established from M2 xenografts and forms high grade carcinomas with the ability to metastasize to the lung(9). This MCF10A series offers the possibility to study the responses of cells with different grades of malignancy that are not biased by a different genetic background. For the analysis of TGF-ß-induced invasion, we generated homotypic MCF10A spheroid cell cultures embedded in a 3D collagen matrix in vitro (Fig 1). Such models closely resemble human tumors in vivo by establishing a gradient of oxygen and nutrients, resulting in active and invasive cells on the outside and quiescent or even necrotic cells in the inside of the spheroid(10). Spheroid based assays have also been shown to better recapitulate drug resistance than monolayer cultures(11). This MCF10 3D model system allowed us to investigate the impact of TGF-ß signaling on the invasive properties of breast cells in different stages of malignancy.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Invasividade Neoplásica , Esferoides Celulares , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia
18.
Biochem Biophys Res Commun ; 412(1): 68-73, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21798242

RESUMO

Amyloid precursor protein (APP), a key protein in pathogenesis of Alzheimer's disease (AD), is a type I transmembrane protein which can be cleaved by ß- and γ-secretase to release the amyloidogenic ß-amyloid peptides (Aß) and the APP intracellular domain (AICD). While Aß has been widely believed to initiate pathogenic cascades culminating AD, the physiological functions and regulations of AICD remain elusive. In present study, endogenous AICD was demonstrated to be increased by canonical Wnt signal. Instead of due to γ-secretase activity, enhanced AICD expression was found due to the increased protein stability by Wnt/ß-catenin. ß-Catenin was demonstrated to be an associating partner of AICD, capable of promoting AICD mediated transcriptional activity. Investigation by AICD mutants proved that Fe65, a previously identified AICD binding partner, is not involved in this regulation. Taken together, our results suggest that AICD is stabilized and the AICD mediated transcriptional activity is promoted by canonical Wnt/ß-catenin signaling independent of Fe65.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Transcrição Gênica , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Células HEK293 , Humanos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Estabilidade Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
19.
Mol Biol Cell ; 22(18): 3533-40, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21795403

RESUMO

Glycogen synthase kinase 3ß (GSK3ß) regulates diverse physiological processes, including metabolism, development, oncogenesis, and neuroprotection. GSK3ß kinase activity has been reported to be critical for various types of cancer cells, but the mechanism has remained elusive. In this study we examine the mechanism by which GSK3ß regulates the survival of leukemia cells. We demonstrate that upon GSK3ß kinase inhibition different types of leukemia cells show severe proliferation defects as a result of apoptosis. The transcription factor c-Myb is found to be the main target of GSK3ß inhibition in cell survival. GSK3ß inactivation reduces the expression of c-Myb by promoting its ubiquitination-mediated degradation, thereby inhibiting the expression of c-Myb-dependent antiapoptotic genes Bcl2 and survivin. Coimmunoprecipitation, reporter assays, chromatin immunoprecipitation, and knockdown studies show that c-Myb needs to interact and cooperate with transcription factor LEF-1 in the activation of Bcl2 and survivin and that both transcription factors are required for cell survival. These data reveal an as-yet-unknown mechanism by which GSK3ß controls cell survival.


Assuntos
Apoptose , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Proto-Oncogênicas c-myb/fisiologia , Proteína Axina/genética , Proteína Axina/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Expressão Gênica , Regulação da Expressão Gênica , Genes Reporter , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Indóis/farmacologia , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leucemia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Maleimidas/farmacologia , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Survivina , Ubiquitinação
20.
Protein Cell ; 2(5): 377-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21626267

RESUMO

Amyloid beta (Aß) precursor protein (APP) is a key protein in the pathogenesis of Alzheimer's disease (AD). Both APP and its paralogue APLP1 (amyloid beta precursor-like protein 1) have multiple functions in cell adhesion and proliferation. Previously it was thought that autophagy is a novel beta-amyloid peptide (Aß)-generating pathway activated in AD. However, the protein proteolysis of APLP1 is still largely unknown. The present study shows that APLP1 is rapidly degraded in neuronal cells in response to stresses, such as proteasome inhibition. Activation of the endoplasmic reticulum (ER) stress by proteasome inhibitors induces autophagy, causing reduction of mature APLP1/APP. Blocking autophagy or JNK stress kinase rescues the protein expression for both APP and APLP1. Therefore, our results suggest that APP/APLP1 is degraded through autophagy and the APLP1 proteolysis is mainly mediated by autophagy-lysosome pathway.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Autofagia , Neurônios/metabolismo , Inibidores de Proteassoma , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leupeptinas/farmacologia , Camundongos , Neurônios/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...