Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 626(8001): 990-998, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38383782

RESUMO

Electrode-based electrical stimulation underpins several clinical bioelectronic devices, including deep-brain stimulators1,2 and cardiac pacemakers3. However, leadless multisite stimulation is constrained by the technical difficulties and spatial-access limitations of electrode arrays. Optogenetics offers optically controlled random access with high spatiotemporal capabilities, but clinical translation poses challenges4-6. Here we show tunable spatiotemporal photostimulation of cardiac systems using a non-genetic platform based on semiconductor-enabled biomodulation interfaces. Through spatiotemporal profiling of photoelectrochemical currents, we assess the magnitude, precision, accuracy and resolution of photostimulation in four leadless silicon-based monolithic photoelectrochemical devices. We demonstrate the optoelectronic capabilities of the devices through optical overdrive pacing of cultured cardiomyocytes (CMs) targeting several regions and spatial extents, isolated rat hearts in a Langendorff apparatus, in vivo rat hearts in an ischaemia model and an in vivo mouse heart model with transthoracic optical pacing. We also perform the first, to our knowledge, optical override pacing and multisite pacing of a pig heart in vivo. Our systems are readily adaptable for minimally invasive clinical procedures using our custom endoscopic delivery device, with which we demonstrate closed-thoracic operations and endoscopic optical stimulation. Our results indicate the clinical potential of the leadless, lightweight and multisite photostimulation platform as a pacemaker in cardiac resynchronization therapy (CRT), in which lead-placement complications are common.


Assuntos
Terapia de Ressincronização Cardíaca , Desenho de Equipamento , Marca-Passo Artificial , Silício , Animais , Camundongos , Ratos , Terapia de Ressincronização Cardíaca/métodos , Endoscopia , Coração , Procedimentos Cirúrgicos Minimamente Invasivos , Isquemia Miocárdica/cirurgia , Isquemia Miocárdica/terapia , Miócitos Cardíacos , Semicondutores , Suínos , Modelos Animais
2.
Nat Methods ; 21(5): 857-867, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38374262

RESUMO

Studies using antigen-presenting systems at the single-cell and ensemble levels can provide complementary insights into T-cell signaling and activation. Although crucial for advancing basic immunology and immunotherapy, there is a notable absence of synthetic material toolkits that examine T cells at both levels, and especially those capable of single-molecule-level manipulation. Here we devise a biomimetic antigen-presenting system (bAPS) for single-cell stimulation and ensemble modulation of T-cell recognition. Our bAPS uses hexapod heterostructures composed of a submicrometer cubic hematite core (α-Fe2O3) and nanostructured silica branches with diverse surface modifications. At single-molecule resolution, we show T-cell activation by a single agonist peptide-loaded major histocompatibility complex; distinct T-cell receptor (TCR) responses to structurally similar peptides that differ by only one amino acid; and the superior antigen recognition sensitivity of TCRs compared with that of chimeric antigen receptors (CARs). We also demonstrate how the magnetic field-induced rotation of hexapods amplifies the immune responses in suspended T and CAR-T cells. In addition, we establish our bAPS as a precise and scalable method for identifying stimulatory antigen-specific TCRs at the single-cell level. Thus, our multimodal bAPS represents a unique biointerface tool for investigating T-cell recognition, signaling and function.


Assuntos
Ativação Linfocitária , Linfócitos T , Linfócitos T/imunologia , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Apresentação de Antígeno , Dióxido de Silício/química , Compostos Férricos/química , Peptídeos/química , Peptídeos/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Nanoestruturas/química , Camundongos , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo
3.
Nat Mater ; 21(6): 647-655, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35618824

RESUMO

Homo- and heterojunctions play essential roles in semiconductor-based devices such as field-effect transistors, solar cells, photodetectors and light-emitting diodes. Semiconductor junctions have been recently used to optically trigger biological modulation via photovoltaic or photoelectrochemical mechanisms. The creation of heterojunctions typically involves materials with different doping or composition, which leads to high cost, complex fabrications and potential side effects at biointerfaces. Here we show that a porosity-based heterojunction, a largely overlooked system in materials science, can yield an efficient photoelectrochemical response from the semiconductor surface. Using self-limiting stain etching, we create a nanoporous/non-porous, soft-hard heterojunction in p-type silicon within seconds under ambient conditions. Upon surface oxidation, the heterojunction yields a strong photoelectrochemical response in saline. Without any interconnects or metal modifications, the heterojunction enables efficient non-genetic optoelectronic stimulation of isolated rat hearts ex vivo and sciatic nerves in vivo with optical power comparable to optogenetics, and with near-infrared capabilities.


Assuntos
Ciência dos Materiais , Semicondutores , Porosidade
4.
Mol Psychiatry ; 26(9): 5266-5276, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33619338

RESUMO

Alcohol use disorder (AUD) is one of the foremost public health problems. Alcohol is also frequently co-abused with cocaine. There is a huge unmet need for the treatment of AUD and/or cocaine co-abuse. We recently demonstrated that skin grafts generated from mouse epidermal stem cells that had been engineered by CRISPR-mediated genome editing could be transplanted onto mice as a gene delivery platform. Here, we show that expression of the glucagon-like peptide-1 (GLP1) gene delivered by epidermal stem cells attenuated development and reinstatement of alcohol-induced drug-taking and seeking as well as voluntary oral alcohol consumption. GLP1 derived from the skin grafts decreased alcohol-induced increase in dopamine levels in the nucleus accumbens. In exploring the potential of this platform in reducing concurrent use of drugs, we developed a novel co-grafting procedure for both modified human butyrylcholinesterase (hBChE)- and GLP1-expressing cells. Epidermal stem cell-derived hBChE and GLP1 reduced acquisition of drug-taking and toxicity induced by alcohol and cocaine co-administration. These results imply that cutaneous gene delivery through skin transplants may add a new option to treat drug abuse and co-abuse.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Animais , Butirilcolinesterase , Camundongos , Núcleo Accumbens , Ratos , Ratos Sprague-Dawley , Recompensa , Autoadministração , Células-Tronco
5.
EMBO Rep ; 21(5): e48566, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32239614

RESUMO

Progenitor cells at the basal layer of skin epidermis play an essential role in maintaining tissue homeostasis and enhancing wound repair in skin. The proliferation, differentiation, and cell death of epidermal progenitor cells have to be delicately regulated, as deregulation of this process can lead to many skin diseases, including skin cancers. However, the underlying molecular mechanisms involved in skin homeostasis remain poorly defined. In this study, with quantitative proteomics approach, we identified an important interaction between KDF1 (keratinocyte differentiation factor 1) and IKKα (IκB kinase α) in differentiating skin keratinocytes. Ablation of either KDF1 or IKKα in mice leads to similar but striking abnormalities in skin development, particularly in skin epidermal differentiation. With biochemical and mouse genetics approach, we further demonstrate that the interaction of IKKα and KDF1 is essential for epidermal differentiation. To probe deeper into the mechanisms, we find that KDF1 associates with a deubiquitinating protease USP7 (ubiquitin-specific peptidase 7), and KDF1 can regulate skin differentiation through deubiquitination and stabilization of IKKα. Taken together, our study unravels an important molecular mechanism underlying epidermal differentiation and skin tissue homeostasis.


Assuntos
Diferenciação Celular , Células Epidérmicas/citologia , Quinase I-kappa B , Queratinócitos , Proteínas/metabolismo , Animais , Epiderme , Quinase I-kappa B/genética , Camundongos , Ubiquitinação
6.
EMBO J ; 36(13): 1963-1980, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28507225

RESUMO

Tissue homeostasis of skin is sustained by epidermal progenitor cells localized within the basal layer of the skin epithelium. Post-translational modification of the proteome, such as protein phosphorylation, plays a fundamental role in the regulation of stemness and differentiation of somatic stem cells. However, it remains unclear how phosphoproteomic changes occur and contribute to epidermal differentiation. In this study, we survey the epidermal cell differentiation in a systematic manner by combining quantitative phosphoproteomics with mammalian kinome cDNA library screen. This approach identified a key signaling event, phosphorylation of a desmosome component, PKP1 (plakophilin-1) by RIPK4 (receptor-interacting serine-threonine kinase 4) during epidermal differentiation. With genome-editing and mouse genetics approach, we show that loss of function of either Pkp1 or Ripk4 impairs skin differentiation and enhances epidermal carcinogenesis in vivo Phosphorylation of PKP1's N-terminal domain by RIPK4 is essential for their role in epidermal differentiation. Taken together, our study presents a global view of phosphoproteomic changes that occur during epidermal differentiation, and identifies RIPK-PKP1 signaling as novel axis involved in skin stratification and tumorigenesis.


Assuntos
Diferenciação Celular , Queratinócitos/fisiologia , Placofilinas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Pele/citologia , Células-Tronco/fisiologia , Animais , Carcinogênese , Células Cultivadas , Perfilação da Expressão Gênica , Camundongos , Camundongos Knockout , Fosforilação , Proteoma/análise , Neoplasias Cutâneas , Transplante de Tecidos
7.
Biochemistry ; 58(27): 2996-3004, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31243996

RESUMO

Anthrax, a lethal, weaponizable disease caused by Bacillus anthracis, acts through exotoxins that are primary mediators of systemic toxicity and also targets for neutralization by passive immunotherapy. The ease of engineering B. anthracis strains resistant to established therapy and the historic use of the microbe in bioterrorism present a compelling test case for platforms that permit the rapid and modular development of neutralizing agents. In vitro antigen-binding fragment (Fab) selection offers the advantages of speed, sequence level molecular control, and engineering flexibility compared to traditional monoclonal antibody pipelines. By screening an unbiased, chemically synthetic phage Fab library and characterizing hits in cell-based assays, we identified two high-affinity neutralizing Fabs, A4 and B7, against anthrax edema factor (EF), a key mediator of anthrax pathogenesis. Engineered homodimers of these Fabs exhibited potency comparable to that of the best reported neutralizing monoclonal antibody against EF at preventing EF-induced cyclic AMP production. Using internalization assays in COS cells, B7 was found to block steps prior to EF internalization. This work demonstrates the efficacy of synthetic alternatives to traditional antibody therapeutics against anthrax while also demonstrating a broadly generalizable, rapid, and modular screening pipeline for neutralizing antibody generation.


Assuntos
Antraz/tratamento farmacológico , Anticorpos Neutralizantes/farmacologia , Bacillus anthracis/efeitos dos fármacos , Toxinas Bacterianas/antagonistas & inibidores , Fragmentos Fab das Imunoglobulinas/farmacologia , Sequência de Aminoácidos , Animais , Antraz/metabolismo , Antraz/microbiologia , Anticorpos Neutralizantes/química , Antígenos de Bactérias/metabolismo , Bacillus anthracis/fisiologia , Toxinas Bacterianas/metabolismo , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetulus , AMP Cíclico/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/química , Camundongos , Multimerização Proteica
8.
J Cell Sci ; 130(15): 2447-2457, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679697

RESUMO

The different cytoskeletal networks in a cell are responsible for many fundamental cellular processes. Current studies have shown that spectraplakins, cytoskeletal crosslinkers that combine features of both the spectrin and plakin families of crosslinkers, have a critical role in integrating these different cytoskeletal networks. Spectraplakin genes give rise to a variety of isoforms that have distinct functions. Importantly, all spectraplakin isoforms are uniquely able to associate with all three elements of the cytoskeleton, namely, F-actin, microtubules and intermediate filaments. In this Review, we will highlight recent studies that have unraveled their function in a wide range of different processes, from regulating cell adhesion in skin keratinocytes to neuronal cell migration. Taken together, this work has revealed a diverse and indispensable role for orchestrating the function of different cytoskeletal elements in vivo.


Assuntos
Citoesqueleto/metabolismo , Filamentos Intermediários/metabolismo , Queratinócitos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Adesão Celular/fisiologia , Humanos , Queratinócitos/citologia , Isoformas de Proteínas/metabolismo
9.
Acta Biochim Biophys Sin (Shanghai) ; 51(5): 501-508, 2019 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-30939192

RESUMO

Elevation of angiotensin II (Ang II) in the serum of patients with diabetes is known to promote apoptosis of islet ß cells, but the underlying mechanism remains unclear. The aim of the present study was to explore the role of Nod-like receptor protein 3 (NLRP3) inflammasome in Ang II-induced apoptosis of pancreatic islet ß cells and investigate the possible underlying mechanism. The effect of Ang II on INS-1 cell (a rat insulinoma cell line) viability was detected by CCK-8 method. The cell apoptosis was detected by flow cytometry and western blot analysis. The effect of Ang II on the expressions of thioredoxin-interacting protein (TXNIP) and NLRP3 protein was detected by western blot analysis. The expression of TXNIP mRNA was detected by real-time polymerase chain reaction. The results showed that Ang II was able to reduce INS-1 cell viability and promote apoptosis and at the same time up-regulate the expressions of TXNIP and NLRP3 components. Ang II-induced apoptosis was inhibited after administration of the NLRP3 inhibitor MCC950, and TXNIP silencing could reduce the NLRP3 expression and apoptosis, while both effects of Ang II on TXNIP-NLRP3 and its apoptosis-inducing effect were inhibited by angiotensin II type I receptor (AT1R) blocker Telmisartan. Our results demonstrated that the TXNIP-NLRP3 inflammasome pathway mediated Ang II-induced INS-1 cell apoptosis and might hopefully become a novel target for the treatment of diabetes mellitus.


Assuntos
Angiotensina II/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Inflamassomos/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Apoptose/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Humanos , Inflamassomos/genética , Células Secretoras de Insulina/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Ratos , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
10.
Nat Mater ; 15(9): 1023-30, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27348576

RESUMO

Silicon-based materials have widespread application as biophysical tools and biomedical devices. Here we introduce a biocompatible and degradable mesostructured form of silicon with multi-scale structural and chemical heterogeneities. The material was synthesized using mesoporous silica as a template through a chemical vapour deposition process. It has an amorphous atomic structure, an ordered nanowire-based framework and random submicrometre voids, and shows an average Young's modulus that is 2-3 orders of magnitude smaller than that of single-crystalline silicon. In addition, we used the heterogeneous silicon mesostructures to design a lipid-bilayer-supported bioelectric interface that is remotely controlled and temporally transient, and that permits non-genetic and subcellular optical modulation of the electrophysiology dynamics in single dorsal root ganglia neurons. Our findings suggest that the biomimetic expansion of silicon into heterogeneous and deformable forms can open up opportunities in extracellular biomaterial or bioelectric systems.

11.
J Biol Chem ; 290(52): 30771-82, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26527684

RESUMO

Cell migration is a fundamental cellular process requiring integrated activities of the cytoskeleton, membrane, and cell/extracellular matrix adhesions. Many cytoskeletal activities rely on microtubule filaments. It has been speculated that microtubules can serve as tracks to deliver proteins essential for focal adhesion turnover. Three microtubule end-binding proteins (EB1, EB2, and EB3) in mammalian cells can track the plus ends of growing microtubules. EB1 and EB3 together can regulate microtubule dynamics by promoting microtubule growth and suppressing catastrophe, whereas, in contrast, EB2 does not play a direct role in microtubule dynamic instability, and little is known about the cellular function of EB2. By quantitative proteomics, we identified mammalian HCLS1-associated protein X-1 (HAX1) as an EB2-specific interacting protein. Knockdown of HAX1 and EB2 in skin epidermal cells stabilizes focal adhesions and impairs epidermal migration in vitro and in vivo. Our results further demonstrate that cell motility and focal adhesion turnover require interaction between Hax1 and EB2. Together, our findings provide new insights for this critical cellular process, suggesting that EB2 association with Hax1 plays a significant role in focal adhesion turnover and epidermal migration.


Assuntos
Adesões Focais/metabolismo , Queratinócitos/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Animais , Movimento Celular , Células Cultivadas , Adesões Focais/genética , Peptídeos e Proteínas de Sinalização Intracelular , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Ligação Proteica , Proteínas/genética
12.
PLoS Pathog ; 10(5): e1004125, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809689

RESUMO

The DOK1 tumor suppressor gene encodes an adapter protein that acts as a negative regulator of several signaling pathways. We have previously reported that DOK1 expression is up-regulated upon cellular stress, via the transcription factor E2F1, and down-regulated in a variety of human malignancies due to aberrant hypermethylation of its promoter. Here we show that Epstein Barr virus (EBV) infection of primary human B-cells leads to the down-regulation of DOK1 gene expression via the viral oncoprotein LMP1. LMP1 alone induces recruitment to the DOK1 promoter of at least two independent inhibitory complexes, one containing E2F1/pRB/DNMT1 and another containing at least EZH2. These events result in tri-methylation of histone H3 at lysine 27 (H3K27me3) of the DOK1 promoter and gene expression silencing. We also present evidence that the presence of additional EBV proteins leads to further repression of DOK1 expression with an additional mechanism. Indeed, EBV infection of B-cells induces DNA methylation at the DOK1 promoter region including the E2F1 responsive elements that, in turn, lose the ability to interact with E2F complexes. Treatment of EBV-infected B-cell-lines with the methyl-transferase inhibitor 5-aza-2'-deoxycytidine rescues DOK1 expression. In summary, our data show the deregulation of DOK1 gene expression by EBV and provide novel insights into the regulation of the DOK1 tumor suppressor in viral-related carcinogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Infecções por Vírus Epstein-Barr/genética , Herpesvirus Humano 4/fisiologia , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética , Linfócitos B/metabolismo , Linfócitos B/virologia , Transformação Celular Viral/genética , Células Cultivadas , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Infecções por Vírus Epstein-Barr/imunologia , Regulação da Expressão Gênica , Inativação Gênica , Genes Supressores de Tumor , Humanos , Fosfoproteínas/metabolismo , Cultura Primária de Células , Proteínas de Ligação a RNA/metabolismo , Proteínas da Matriz Viral/fisiologia
14.
Science ; 384(6699): 1023-1030, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38815037

RESUMO

Seamless interfaces between electronic devices and biological tissues stand to revolutionize disease diagnosis and treatment. However, biological and biomechanical disparities between synthetic materials and living tissues present challenges at bioelectrical signal transduction interfaces. We introduce the active biointegrated living electronics (ABLE) platform, encompassing capabilities across the biogenic, biomechanical, and bioelectrical properties simultaneously. The living biointerface, comprising a bioelectronics layout and a Staphylococcus epidermidis-laden hydrogel composite, enables multimodal signal transduction at the microbial-mammalian nexus. The extracellular components of the living hydrogels, prepared through thermal release of naturally occurring amylose polymer chains, are viscoelastic, capable of sustaining the bacteria with high viability. Through electrophysiological recordings and wireless probing of skin electrical impedance, body temperature, and humidity, ABLE monitors microbial-driven intervention in psoriasis.


Assuntos
Hidrogéis , Psoríase , Pele , Staphylococcus epidermidis , Animais , Humanos , Camundongos , Temperatura Corporal , Impedância Elétrica , Eletrônica , Umidade , Hidrogéis/química , Inflamação/microbiologia , Inflamação/terapia , Pele/microbiologia , Dispositivos Eletrônicos Vestíveis , Tecnologia sem Fio , Psoríase/microbiologia , Psoríase/terapia , Camundongos Knockout , Receptor 2 Toll-Like/genética
15.
Sci Adv ; 9(29): eadg5858, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37478187

RESUMO

Semiconductor-based biointerfaces are typically established either on the surface of the plasma membrane or within the cytoplasm. In Gram-negative bacteria, the periplasmic space, characterized by its confinement and the presence of numerous enzymes and peptidoglycans, offers additional opportunities for biomineralization, allowing for nongenetic modulation interfaces. We demonstrate semiconductor nanocluster precipitation containing single- and multiple-metal elements within the periplasm, as observed through various electron- and x-ray-based imaging techniques. The periplasmic semiconductors are metastable and display defect-dominant fluorescent properties. Unexpectedly, the defect-rich (i.e., the low-grade) semiconductor nanoclusters produced in situ can still increase adenosine triphosphate levels and malate production when coupled with photosensitization. We expand the sustainability levels of the biohybrid system to include reducing heavy metals at the primary level, building living bioreactors at the secondary level, and creating semi-artificial photosynthesis at the tertiary level. The biomineralization-enabled periplasmic biohybrids have the potential to serve as defect-tolerant platforms for diverse sustainable applications.


Assuntos
Biomineralização , Periplasma , Periplasma/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Fotossíntese
16.
Nat Chem ; 15(1): 119-128, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36280766

RESUMO

Interactions between the microbiota and their colonized environments mediate critical pathways from biogeochemical cycles to homeostasis in human health. Here we report a soil-inspired chemical system that consists of nanostructured minerals, starch granules and liquid metals. Fabricated via a bottom-up synthesis, the soil-inspired chemical system can enable chemical redistribution and modulation of microbial communities. We characterize the composite, confirming its structural similarity to the soil, with three-dimensional X-ray fluorescence and ptychographic tomography and electron microscopy imaging. We also demonstrate that post-synthetic modifications formed by laser irradiation led to chemical heterogeneities from the atomic to the macroscopic level. The soil-inspired material possesses chemical, optical and mechanical responsiveness to yield write-erase functions in electrical performance. The composite can also enhance microbial culture/biofilm growth and biofuel production in vitro. Finally, we show that the soil-inspired system enriches gut bacteria diversity, rectifies tetracycline-induced gut microbiome dysbiosis and ameliorates dextran sulfate sodium-induced rodent colitis symptoms within in vivo rodent models.


Assuntos
Colite , Microbioma Gastrointestinal , Humanos , Animais , Solo/química , Colite/induzido quimicamente , Colite/metabolismo , Homeostase , Modelos Animais de Doenças
17.
Int J Cancer ; 130(11): 2484-94, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21796618

RESUMO

The DOK1 gene is a putative tumour suppressor gene located on the human chromosome 2p13 which is frequently rearranged in leukaemia and other human tumours. We previously reported that the DOK1 gene can be mutated and its expression down-regulated in human malignancies. However, the mechanism underlying DOK1 silencing remains largely unknown. We show here that unscheduled silencing of DOK1 expression through aberrant hypermethylation is a frequent event in a variety of human malignancies. DOK1 was found to be silenced in nine head and neck cancer (HNC) cell lines studied and DOK1 CpG hypermethylation correlated with loss of gene expression in these cells. DOK1 expression could be restored via demethylating treatment using 5-aza-2'deoxycytidine. In addition, transduction of cancer cell lines with DOK1 impaired their proliferation, consistent with the critical role of epigenetic silencing of DOK1 in the development and maintenance of malignant cells. We further observed that DOK1 hypermethylation occurs frequently in a variety of primary human neoplasm including solid tumours (93% in HNC, 81% in lung cancer) and haematopoietic malignancy (64% in Burkitt's lymphoma). Control blood samples and exfoliated mouth epithelial cells from healthy individuals showed a low level of DOK1 methylation, suggesting that DOK1 hypermethylation is a tumour specific event. Finally, an inverse correlation was observed between the level of DOK1 gene methylation and its expression in tumour and adjacent non tumour tissues. Thus, hypermethylation of DOK1 is a potentially critical event in human carcinogenesis, and may be a potential cancer biomarker and an attractive target for epigenetic-based therapy.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/genética , Neoplasias de Cabeça e Pescoço/genética , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA/genética , Adulto , Idoso , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Decitabina , Feminino , Genes Supressores de Tumor , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/antagonistas & inibidores , Proteínas de Ligação a RNA/antagonistas & inibidores , Fatores de Risco , Proteínas Supressoras de Tumor/genética
18.
J Virol ; 85(17): 9013-22, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21715489

RESUMO

Constitutive activation of NF-κB signaling is a key event in virus- and non-virus-induced carcinogenesis. We have previously reported that cutaneous human papillomavirus type 38 (HPV38) displays transforming properties in in vitro and in vivo experimental models. However, the involvement of NF-κB signaling in HPV38-induced cell growth transformation remains to be determined. In this study, we showed that HPV38 E6 and E7 activate NF-κB and that inhibition of the pathway with the IκBα superrepressor sensitizes HPV38E6E7-immortalized human keratinocytes to tumor necrosis factor alpha (TNF-α)- and UVB radiation-mediated apoptosis. Accordingly, inhibition of NF-κB signaling resulted in the downregulation of NF-κB-regulated antiapoptotic genes, including cIAP1, cIAP2, and xIAP genes. These findings demonstrate a critical role of NF-κB activity in the survival of HPV38E6E7-immortalized human keratinocytes exposed to cytokine or UV radiation. Our data provide additional evidence for cooperation between beta HPV infection and UV irradiation in skin carcinogenesis.


Assuntos
Apoptose , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , NF-kappa B/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/patogenicidade , Fator de Necrose Tumoral alfa/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos
19.
J Virol ; 85(17): 8477-94, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21697493

RESUMO

We previously reported that the oncoproteins E6 and E7 from cutaneous human papillomavirus type 38 (HPV38) can immortalize primary human keratinocytes in vitro and sensitize transgenic mice to develop skin cancer in vivo. Immunofluorescence staining revealed that human keratinocytes immortalized by HPV38 E6 and E7 display fewer actin stress fibers than do control primary keratinocyte cells, raising the possibility of a role of the viral oncoproteins in the remodeling of the actin cytoskeleton. In this study, we show that HPV38 E7 induces actin stress fiber disruption and that this phenomenon correlates with its ability to downregulate Rho activity. The downregulation of Rho activity by HPV38 E7 is mediated through the activation of the CK2-MEK-extracellular signal-regulated kinase (ERK) pathway. In addition, HPV38 E7 is able to induce actin fiber disruption by binding directly to eukaryotic elongation factor 1A (eEF1A) and abolishing its effects on actin fiber formation. Finally, we found that the downregulation of Rho activity by HPV38 E7 through the CK2-MEK-ERK pathway facilitates cell growth proliferation. Taken together, our data support the conclusion that HPV38 E7 promotes keratinocyte proliferation in part by negatively regulating actin cytoskeleton fiber formation through the CK2-MEK-ERK-Rho pathway and by binding to eEF1A and inhibiting its effects on actin cytoskeleton remodeling.


Assuntos
Actinas/metabolismo , Caseína Quinase II/metabolismo , Citoesqueleto/metabolismo , Fator de Iniciação 1 em Eucariotos/antagonistas & inibidores , Queratinócitos/virologia , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/patogenicidade , Linhagem Celular , Proliferação de Células , Humanos , Ligação Proteica
20.
Mol Pharm ; 9(11): 3259-65, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23009653

RESUMO

Owing to advantageous biochemical and pharmacological properties of human serum albumin (HSA), HSA-based drug carrier is playing an increasing role in the clinical setting. Since the IIA subdomain of HSA is a big hydrophobic cavity, we proposed that HSA delivers multiple drugs simultaneously docking on the IIA subdomain and that drugs may influence each other's binding affinity to albumin when cobinding the HSA IIA subdomain. Therefore, we studied the interactive association of drugs with the IIA subdomain of HSA by fluorescence spectroscopy and X-ray crystallography, in order to elucidate the molecular mechanism and behavior of drugs cobinding the IIA subdomain of HSA, and develop a universal structure-based model for HSA carrier design. We solved HSA-fatty acid-cinnamic acid, HSA-fatty acid-cinnamic acid-indometacin and HSA-fatty acid-cinnamic acid-indometacin-lamivudine complex structures, respectively. HSA complex structures and fluorescence quenching of HSA revealed that different drugs can regulate binding sites, binding mode and binding affinity of each other. For example, indometacin renders cinnamic acid to make reposition, and decreases binding affinity of HSA for cinnamic acid, but increases binding affinity of itself to HSA. Lamivudine makes cinnamic acid and indometacin to bind new subsites. Cinnamic acid-indometacin enhances binding affinity of lamivudine, and cinnamic acid-lamivudine increases binding affinity of indometacin, but indometacin-lamivudine decreases binding affinity of cinnamic acid to HSA. The study provided a biochemical basis for structure-guided development of HSA delivery system.


Assuntos
Cinamatos/metabolismo , Ácidos Graxos/metabolismo , Indometacina/metabolismo , Lamivudina/metabolismo , Albumina Sérica/metabolismo , Sítios de Ligação , Cinamatos/química , Cristalografia por Raios X , Ácidos Graxos/química , Humanos , Indometacina/química , Lamivudina/química , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , Albumina Sérica/química , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA