Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; : e2312219, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38608672

RESUMO

Targeting the competitive-cooperative relationships among tumor cells and various immune cells can efficiently reverse the immune-dysfunction microenvironment to boost the immunotherapies for the triple-negative breast cancer treatment. Hence, a bacterial outer membrane vesicle-based nanocomplex is designed for specifically targeting malignant cells and immune cells to reconcile the relationships based on metabolic-immune crosstalk. By uniquely utilizing the property of charge-reversal polymers to realize function separation, the nanocomplexes could synergistically regulate tumor cells and immune cells. This approach could reshape the immunosuppressive competition-cooperation pattern into one that is immune-responsive, showcasing significant potential for inducing tumor remission in TNBC models.

2.
Biophys J ; 122(24): 4656-4669, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-37974397

RESUMO

Serine integrases promote the recombination of two complementary DNA sequences, attP and attB, to create hybrid sequences, attL and attR. The reaction is unidirectional in the absence of an accessory protein called recombination directionality factor. We utilized tethered particle motion (TPM) experiments to investigate the reaction behaviors of two model serine integrases from Listeria innocua phage LI and Streptomyces coelicolor phage C31. Detailed kinetic analyses of wild-type and mutant proteins were carried out to verify the mechanisms of recombination directionality. In particular, we assessed the influence of a coiled-coil motif (CC) that is conserved in the C-terminal domain of serine integrases and is an important prerequisite for efficient recombination. Compared to wild type, we found that CC deletions in both serine integrases reduced the overall abundance of integrase (Int) att-site complexes and favored the formation of nonproductive complexes over recombination-competent complexes. Furthermore, the rate at which CC mutants formed productive synaptic complexes and disassembled aberrant nonproductive complexes was significantly reduced. It is notable that while the φC31 Int CC is essential for recombination, the LI Int CC plays an auxiliary role for recombination to stabilize protein-protein interactions and to control the directionality of the reaction.


Assuntos
Bacteriófagos , Recombinases , Recombinases/genética , Serina/metabolismo , Sítios de Ligação Microbiológicos , Recombinação Genética , Integrases/genética , Integrases/metabolismo , Bacteriófagos/genética
3.
ACS Nano ; 17(18): 18164-18177, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37703316

RESUMO

Reperfusion injury presents a significant obstacle to neuronal survival following successful recanalization in ischemic stroke, which is characterized by intricate pathophysiological processes comprising numerous interconnected pathways. Oxidative stress-induced neuronal ferroptosis and the overactivation of glial cells play important roles in this phenomenon. In this study, we developed a targeted cross-linked micelle loaded with idebenone to rescue the ischemic penumbra by inhibiting neuronal ferroptosis and glial overactivation. In rat models, the CREKA peptide-modified micelles accumulate in the damaged brain via binding to microthrombi in the ipsilateral microvessels. Upon reactive oxygen species (ROS) stimulation, diselenide bonds within the micelles are transformed to hydrophilic seleninic acids, enabling synchronized ROS consumption and responsive drug release. The released idebenone scavenges ROS, prevents oxidative stress-induced neuronal ferroptosis, attenuates glial overactivation, and suppresses pro-inflammatory factors secretion, thereby modulating the inflammatory microenvironment. Finally, this micelle significantly reinforces neuronal survival, reduces infarct volume, and improves behavioral function compared to the control groups. This pleiotropic therapeutic micelle provides a proof-of-concept of remodeling the lesion microenvironment by inhibiting neuronal ferroptosis and glial overactivation to treat cerebral ischemia-reperfusion injury.


Assuntos
Ferroptose , Traumatismo por Reperfusão , Animais , Ratos , Micelas , Espécies Reativas de Oxigênio , Neuroglia , Traumatismo por Reperfusão/tratamento farmacológico
4.
Acta Biomater ; 167: 387-400, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37276955

RESUMO

The tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) is the main block for the penetration of chemotherapy. In the tumor microenvironment, a dense matrix composed of fibrin is formed on the exterior, while the interior is featured by high reduction, hypoxia and low pH. How to match the special microenvironment to on-demand drug release is the key to improve chemotherapeutic efficacy. Herein, a microenvironment-responsive micellar system is developed to deepen tumoral penetration. Briefly, the conjugation of a fibrin-targeting peptide to PEG-poly amino acid has been utilized to achieve accumulation of micelles in the tumor stroma. By modification of micelles with hypoxia-reducible nitroimidazole which becomes protonated under acidic conditions, their surface charge is more positive, facilitating deeper penetration into tumors. Paclitaxel was loaded onto the micelles via a disulfide bond to enable glutathione (GSH)-responsive release. Therefore, the immunosuppressive microenvironment is relived through the alleviation of hypoxia and depletion of GSH. Hopefully, this work could establish paradigms by designing sophisticated drug-delivery systems to tactfully employ and retroact the tamed tumoral microenvironment to improve the therapeutic efficacy based on understanding the multiple hallmarks and learning the mutual regulation. STATEMENT OF SIGNIFICANCE: Tumor microenvironment(TME) is an unique pathological feature of pancreatic cancer and an inherent barrier to chemotherapy. Numerous studies regard TME as the targets for drug delivery. In this work, we propose a hypoxia-responsive nanomicellar drug delivery system that aiming hypoxia TME of pancreatic cancer. The nanodrug delivery system could respond to the hypoxic microenvironment and enhance the penetration of the inner tumor at the same time preserving the outer tumor stroma, thus achieving targeted treatment of PDAC by preserving the integrity of the outer stroma. Simultaneously, the responsive group can reverse the degree of hypoxia in TME by disrupting the redox balance in the tumor region, thus achieving precise treatment of PDAC by matching the pathological characteristics of TME. We believe our article would provide new design ideas for the future treatments for pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Micelas , Microambiente Tumoral , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Hipóxia , Glutationa , Terapia de Imunossupressão , Linhagem Celular Tumoral , Neoplasias Pancreáticas
5.
Bioact Mater ; 27: 474-487, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37159612

RESUMO

A long-standing paucity of effective therapies results in the poor outcomes of triple-negative breast cancer brain metastases. Immunotherapy has made progress in the treatment of tumors, but limited by the non-immunogenicity of tumors and strong immunosuppressive environment, patients with TNBC brain metastases have not yet benefited from immunotherapy. Dual immunoregulatory strategies with enhanced immune activation and reversal of the immunosuppressive microenvironment provide new therapeutic options for patients. Here, we propose a cocktail-like therapeutic strategy of microenvironment regulation-chemotherapy-immune synergistic sensitization and construct reduction-sensitive immune microenvironment regulation nanomaterials (SIL@T). SIL@T modified with targeting peptide penetrates the BBB and is subsequently internalized into metastatic breast cancer cells, releasing silybin and oxaliplatin responsively in the cells. SIL@T preferentially accumulates at the metastatic site and can significantly prolong the survival period of model animals. Mechanistic studies have shown that SIL@T can effectively induce immunogenic cell death of metastatic cells, activate immune responses and increase infiltration of CD8+ T cells. Meanwhile, the activation of STAT3 in the metastatic foci is attenuated and the immunosuppressive microenvironment is reversed. This study demonstrates that SIL@T with dual immunomodulatory functions provides a promising immune synergistic therapy strategy for breast cancer brain metastases.

6.
Acta Pharm Sin B ; 13(1): 298-314, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36815033

RESUMO

Metastasis accounts for 90% of breast cancer deaths, where the lethality could be attributed to the poor drug accumulation at the metastatic loci. The tolerance to chemotherapy induced by breast cancer stem cells (BCSCs) and their particular redox microenvironment further aggravate the therapeutic dilemma. To be specific, therapy-resistant BCSCs can differentiate into heterogeneous tumor cells constantly, and simultaneously dynamic maintenance of redox homeostasis promote tumor cells to retro-differentiate into stem-like state in response to cytotoxic chemotherapy. Herein, we develop a specifically-designed biomimic platform employing neutrophil membrane as shell to inherit a neutrophil-like tumor-targeting capability, and anchored chemotherapeutic and BCSCs-differentiating reagents with nitroimidazole (NI) to yield two hypoxia-responsive prodrugs, which could be encapsulated into a polymeric nitroimidazole core. The platform can actively target the lung metastasis sites of triple negative breast cancer (TNBC), and release the escorted drugs upon being triggered by the hypoxia microenvironment. During the responsiveness, the differentiating agent could promote transferring BCSCs into non-BCSCs, and simultaneously the nitroimidazole moieties conjugated on the polymer and prodrugs could modulate the tumor microenvironment by depleting nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) and amplifying intracellular oxidative stress to prevent tumor cells retro-differentiation into BCSCs. In combination, the BCSCs differentiation and tumor microenvironment modulation synergistically could enhance the chemotherapeutic cytotoxicity, and remarkably suppress tumor growth and lung metastasis. Hopefully, this work can provide a new insight in to comprehensively treat TNBC and lung metastasis using a versatile platform.

7.
Mol Pharm ; 20(3): 1591-1598, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36715483

RESUMO

Breast cancer stem cells (BCSCs) are the culprit of triple-negative breast cancer invasiveness and are heterogeneous. It is recognized that the combination of chemotherapy and differentiation therapy for killing BCSCs and non-BCSCs simultaneously is a reliable strategy. In this study, an oil-in-water nanoemulsion was prepared by high-pressure homogenization with coencapsulation of all-trans retinoic acid (ATRA) and doxorubicin (DOX). The preparation process was simple, and the production was easy to scale up. The particle size of the nanoemulsion was 127.2 ± 2.0 nm. Cellular toxicity assay showed that the composite index of the ATRA and DOX was less than 1 and exhibited a fine combined effect. In vivo antitumor efficacy showed that the compound nanoemulsion could reduce the proportion of BCSCs to 1.18% by inhibiting the expression of Pin1. In addition, the combination of ATRA and DOX could reduce the cardiotoxicity of DOX and had higher safety. Hopefully, this work can provide a new insight into developing pharmaceutically acceptable technology for treating BCSCs.


Assuntos
Antineoplásicos , Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Tretinoína , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Diferenciação Celular , Linhagem Celular Tumoral , Peptidilprolil Isomerase de Interação com NIMA
8.
Biomaterials ; 287: 121599, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35777332

RESUMO

The compact extracellular matrix (ECM) of pancreatic ductal adenocarcinoma (PDAC) is the major physical barrier that hinders the delivery of anti-tumor drugs, leading to strong inherent chemotherapy resistance as well as establishing an immunosuppressive tumor microenvironment (TME). However, forcibly destroying the stroma barrier would break the balance of delicate signal transduction and dependence between tumor cells and matrix components. Uncontrollable growth and metastasis would occur, making PDAC more difficult to control. Hence, we design and construct an aptamer-decorated hypoxia-responsive nanoparticle s(DGL)n@Apt co-loading gemcitabine monophosphate and STAT3 inhibitor HJC0152. This nanoparticle can reverse its surficial charge in the TME, and reduce the size triggered by hypoxia. The released ultra-small DGL particles loading gemcitabine monophosphate exhibit excellent deep-tumor penetration, chemotherapy drugs endocytosis promotion, and autophagy induction ability. Meanwhile, HJC0152 inhibits overactivated STAT3 in both tumor cells and tumor stroma, softens the stroma barrier, and reeducates the TME into an immune-activated state. This smart codelivery strategy provides an inspiring opportunity in PDAC treatment.

9.
Small ; 18(18): e2107712, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35285149

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is on of the most lethal malignant tumors with relatively poor prognosis, characterized with insufficient drug penetration, low immune response and obvious drug resistances. The therapeutic inefficiency is multifactorially related to its specific tumor microenvironment (TME), which is representatively featured as rich stroma and immunosuppression. In this work, a versatile drug delivery system is developed that can coencapsulate two prodrugs modified from gemcitabine (GEM) and a signal transducer and activator of transcription 3 (STAT3) inhibitor (HJC0152), and the gradient pH variation is further sensed in the TME of PDAC to achieve a higher penetration by reversing its surficial charges. The escorted prodrugs can release GEM intracellularly, and respond to the hypoxic condition to yield the parental STAT3 inhibitor HJC0152, respectively. By inhibiting STAT3, the tumor immunosuppression microenvironment can be re-educated through the reversion of M2-like tumor associated macrophages (M2-TAMs), recruitment of cytotoxic T lymphocytes and downregulation of regulatory T cells (Treg s). Furthermore, cytidine deaminase (CDA) and α-smooth muscle actin (α-SMA) expression can be downregulated, plus the lipid modification of GEM, the drug resistance of GEM can be greatly relieved. Based on the above design, a synergetic therapeutic efficacy in PDAC treatment can be achieved to provide more opportunity for clinical applications.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Pró-Fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Resistência a Medicamentos , Humanos , Terapia de Imunossupressão , Micelas , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Pró-Fármacos/uso terapêutico , Microambiente Tumoral , Neoplasias Pancreáticas
10.
Adv Healthc Mater ; 11(3): e2101578, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34800085

RESUMO

The rapid postoperative recurrence and short survival time of glioblastoma (GBM) patients necessitate immediate and effective postoperative treatment. Herein, an immediate and mild postoperative local treatment strategy is developed that regulates the postoperative microenvironment and delays GBM recurrence. Briefly, an injectable hydrogel system (imGEL) loaded with Zn(II)2 -AMD3100 (AMD-Zn) and CpG oligonucleotide nanoparticles (CpG NPs) is injected into the operation cavity, with long-term function to block the recruitment of microglia/ macrophages and activate cytotoxic T cells. The finding indicated that the imGEL can regulate the immune microenvironment, inhibit GBM recurrence, and gain valuable time for subsequent adjuvant clinical chemotherapy.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/cirurgia , Humanos , Hidrogéis/uso terapêutico , Nanopartículas/uso terapêutico , Microambiente Tumoral
11.
ACS Nano ; 15(8): 13826-13838, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34382768

RESUMO

Metabolic interactions between different cell types in the tumor microenvironment (TME) often result in reprogramming of the metabolism to be totally different from their normal physiological processes in order to support tumor growth. Many studies have attempted to inhibit tumor growth and activate tumor immunity by regulating the metabolism of tumors and other cells in TME. However, metabolic inhibitors often suffer from the heterogeneity of tumors, since the favorable metabolic regulation of malignant cells and other cells in TME is often inconsistent with each other. Therefore, we reported the design of a pH-sensitive drug delivery system that targets different cells in TME successively. Outer membrane vesicles (OMVs) derived from Gram-negative bacteria were applied to coload paclitaxel (PTX) and regulated in development and DNA damage response 1 (Redd1)-siRNA and regulate tumor metabolism microenvironment and suppress tumor growth. Our siRNA@M-/PTX-CA-OMVs could first release PTX triggered by the tumor pH (pH 6.8). Then the rest of it would be taken in by M2 macrophages to increase their level of glycolysis. Great potential was observed in TAM repolarization, tumor suppression, tumor immune activation, and TME remolding in the triple-negative breast cancer model. The application of the OMV provided an insight for establishing a codelivery platform for chemical drugs and genetic medicines.


Assuntos
Membrana Externa Bacteriana , Vesículas Extracelulares , RNA Interferente Pequeno/metabolismo , Macrófagos/metabolismo , Bactérias Gram-Negativas , Microambiente Tumoral
12.
Adv Sci (Weinh) ; 8(20): e2101526, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34436822

RESUMO

Reperfusion injury is still a major challenge that impedes neuronal survival in ischemic stroke. However, the current clinical treatments are remained on single pathological process, which are due to lack of comprehensive neuroprotective effects. Herein, a macrophage-disguised honeycomb manganese dioxide (MnO2 ) nanosphere loaded with fingolimod (FTY) is developed to salvage the ischemic penumbra. In particular, the biomimetic nanoparticles can accumulate actively in the damaged brain via macrophage-membrane protein-mediated recognition with cell adhesion molecules that are overexpressed on the damaged vascular endothelium. MnO2 nanosphere can consume excess hydrogen peroxide (H2 O2 ) and convert it into desiderated oxygen (O2 ), and can be decomposed in acidic lysosome for cargo release, so as to reduce oxidative stress and promote the transition of M1 microglia to M2 type, eventually reversing the proinflammatory microenvironment and reinforcing the survival of damaged neuron. This biomimetic nanomedicine raises new strategy for multitargeted combined treatment of ischemic stroke.


Assuntos
Inflamação/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Nanopartículas/química , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Microambiente Celular/efeitos dos fármacos , Cloridrato de Fingolimode/química , Cloridrato de Fingolimode/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , AVC Isquêmico/genética , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/genética , Macrófagos/efeitos dos fármacos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Nanosferas/química , Neurônios/patologia , Neuroproteção , Óxidos/química , Óxidos/farmacologia , Oxigênio/metabolismo , Cultura Primária de Células , Ratos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
13.
Adv Sci (Weinh) ; 8(20): e2102256, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34398516

RESUMO

A versatile tumor-targeting stimuli-responsive theranostic platform for peritoneal metastases of colorectal cancer is proposed in this work for tumor tracking and photothermal-enhanced chemotherapy. A quenched photosensitizer ("off" state) is developed and escorted into a tumor-targeting oxaliplatin-embedded micelle. Once reaching the tumor cell, the micelle is clasped to release free oxaliplatin, as well as the "off" photosensitizer, which is further activated ("turned-on") in the tumor reducing microenvironment to provide optical imaging and photothermal effect. The combined results from hyperthermia-enhanced chemotherapy, deep penetration, perfused O2 , and the leveraged GSH-ROS imbalance in tumor cells are achieved for improved antitumor efficacy and reduced systematic toxicity.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Tratamento Farmacológico , Oxaliplatina/farmacologia , Neoplasias Peritoneais/tratamento farmacológico , Terapia Fototérmica , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Humanos , Camundongos , Metástase Neoplásica , Oxaliplatina/química , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Medicina de Precisão , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Adv Mater ; 33(26): e2100746, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33998706

RESUMO

Current therapeutic strategies for Alzheimer's disease (AD) treatments mainly focus on ß-amyloid (Aß) targeting. However, such therapeutic strategies have limited clinical outcomes due to the chronic and irreversible impairment of the nervous system in the late stage of AD. Recently, inflammatory responses, manifested in oxidative stress and glial cell activation, have been reported as hallmarks in the early stages of AD. Based on the crosstalk between inflammatory response and brain cells, a reactive oxygen species (ROS)-responsive dendrimer-peptide conjugate (APBP) is devised to target the AD microenvironment and inhibit inflammatory responses at an early stage. With the modification of the targeting peptide, this nanoconjugate can efficiently deliver peptides to the infected regions and restore the antioxidant ability of neurons by activating the nuclear factor (erythroid-derived 2)-like 2 signaling pathway. Moreover, this multi-target strategy exhibits a synergistic function of ROS scavenging, promoting Aß phagocytosis, and normalizing the glial cell phenotype. As a result, the nanoconjugate can reduce ROS level, decrease Aß burden, alleviate glial cell activation, and eventually enhance cognitive functions in APPswe/PSEN1dE9 model mice. These results indicate that APBP can be a promising candidate for the multi-target treatment of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Biomimética , Dendrímeros , Camundongos , Microglia , Neurônios/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
15.
IEEE J Biomed Health Inform ; 25(9): 3396-3407, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33945489

RESUMO

Non-invasive heart rate estimation is of great importance in daily monitoring of cardiovascular diseases. In this paper, a bidirectional long short term memory (bi-LSTM) regression network is developed for non-invasive heart rate estimation from the ballistocardiograms (BCG) signals. The proposed deep regression model provides an effective solution to the existing challenges in BCG heart rate estimation, such as the mismatch between the BCG signals and ground-truth reference, multi-sensor fusion and effective time series feature learning. Allowing label uncertainty in the estimation can reduce the manual cost of data annotation while further improving the heart rate estimation performance. Compared with the state-of-the-art BCG heart rate estimation methods, the strong fitting and generalization ability of the proposed deep regression model maintains better robustness to noise (e.g., sensor noise) and perturbations (e.g., body movements) in the BCG signals and provides a more reliable solution for long term heart rate monitoring.


Assuntos
Balistocardiografia , Curadoria de Dados , Frequência Cardíaca , Humanos , Monitorização Fisiológica , Movimento
16.
Nucleic Acids Res ; 48(12): 6413-6430, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32479633

RESUMO

Streptomyces phage ϕC31 integrase (Int)-a large serine site-specific recombinase-is autonomous for phage integration (attP x attB recombination) but is dependent on the phage coded gp3, a recombination directionality factor (RDF), for prophage excision (attL x attR recombination). A previously described activating mutation, E449K, induces Int to perform attL x attR recombination in the absence of gp3, albeit with lower efficiency. E449K has no adverse effect on the competence of Int for attP x attB recombination. Int(E449K) resembles Int in gp3 mediated stimulation of attL x attR recombination and inhibition of attP x attB recombination. Using single-molecule analyses, we examined the mechanism by which E449K activates Int for gp3-independent attL x attR recombination. The contribution of E449K is both thermodynamic and kinetic. First, the mutation modulates the relative abundance of Int bound attL-attR site complexes, favoring pre-synaptic (PS) complexes over non-productively bound complexes. Roughly half of the synaptic complexes formed from Int(E449K) pre-synaptic complexes are recombination competent. By contrast, Int yields only inactive synapses. Second, E449K accelerates the dissociation of non-productively bound complexes and inactive synaptic complexes formed by Int. The extra opportunities afforded to Int(E499K) in reattempting synapse formation enhances the probability of success at fruitful synapsis.


Assuntos
Mutação com Ganho de Função , Integrases/metabolismo , Siphoviridae/enzimologia , Proteínas Virais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Integrases/química , Integrases/genética , Cinética , Simulação de Dinâmica Molecular , Ligação Proteica , Recombinação Genética , Siphoviridae/genética , Proteínas Virais/química , Proteínas Virais/genética
17.
PLoS Genet ; 15(6): e1008193, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31242181

RESUMO

Mechanisms for highly efficient chromosome-associated equal segregation, and for maintenance of steady state copy number, are at the heart of the evolutionary success of the 2-micron plasmid as a stable multi-copy extra-chromosomal selfish DNA element present in the yeast nucleus. The Flp site-specific recombination system housed by the plasmid, which is central to plasmid copy number maintenance, is regulated at multiple levels. Transcription of the FLP gene is fine-tuned by the repressor function of the plasmid-coded partitioning proteins Rep1 and Rep2 and their antagonist Raf1, which is also plasmid-coded. In addition, the Flp protein is regulated by the host's post-translational modification machinery. Utilizing a Flp-SUMO fusion protein, which functionally mimics naturally sumoylated Flp, we demonstrate that the modification signals ubiquitination of Flp, followed by its proteasome-mediated degradation. Furthermore, reduced binding affinity and cooperativity of the modified Flp decrease its association with the plasmid FRT (Flp recombination target) sites, and/or increase its dissociation from them. The resulting attenuation of strand cleavage and recombination events safeguards against runaway increase in plasmid copy number, which is deleterious to the host-and indirectly-to the plasmid. These results have broader relevance to potential mechanisms by which selfish genomes minimize fitness conflicts with host genomes by holding in check the extra genetic load they pose.


Assuntos
DNA Nucleotidiltransferases/genética , Sequências Repetitivas de Ácido Nucleico/genética , Proteína SUMO-1/genética , Transcrição Gênica , Segregação de Cromossomos/genética , Variações do Número de Cópias de DNA/genética , Replicação do DNA/genética , Genoma Fúngico/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ligação Proteica/genética , Processamento de Proteína Pós-Traducional/genética , Proteínas Proto-Oncogênicas c-raf/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Sumoilação/genética , Transativadores/genética
18.
Biochim Biophys Acta Gene Regul Mech ; 1862(2): 129-140, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30593928

RESUMO

Eukaryotes have evolved a specific strategy to package DNA. The nucleosome is a 147-base-pair DNA segment wrapped around histone core proteins that plays important roles regulating DNA-dependent biosynthesis and gene expression. Chromatin remodeling complexes (RSC, Remodel the Structure of Chromatin) hydrolyze ATP to perturb DNA-histone contacts, leading to nucleosome sliding and ejection. Here, we utilized tethered particle motion (TPM) experiments to investigate the mechanism of RSC-mediated nucleosome remodeling in detail. We observed ATP-dependent RSC-mediated DNA looping and nucleosome ejection along individual mononucleosomes and dinucleosomes. We found that nucleosome assembly protein 1 (Nap1) enhanced RSC-mediated nucleosome ejection in a two-step disassembly manner from dinucleosomes but not from mononucleosomes. Based on this work, we provide an entire reaction scheme for the RSC-mediated nucleosome remodeling process that includes DNA looping, nucleosome ejection, the influence of adjacent nucleosomes, and the coordinated action between Nap1 and RSC.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/metabolismo , Eucariotos/genética , Proteína 1 de Modelagem do Nucleossomo/metabolismo , Nucleossomos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Trifosfato de Adenosina/metabolismo , DNA/metabolismo , Histonas/metabolismo
19.
IEEE Trans Biomed Eng ; 65(11): 2634-2648, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29993384

RESUMO

A multiple instance dictionary learning approach, dictionary learning using functions of multiple instances (DL-FUMI), is used to perform beat-to-beat heart rate estimation and to characterize heartbeat signatures from ballistocardiogram (BCG) signals collected with a hydraulic bed sensor. DL-FUMI estimates a "heartbeat concept" that represents an individual's personal ballistocardiogram heartbeat pattern. DL-FUMI formulates heartbeat detection and heartbeat characterization as a multiple instance learning problem to address the uncertainty inherent in aligning BCG signals with ground truth during training. Experimental results show that the estimated heartbeat concept obtained by DL-FUMI is an effective heartbeat prototype and achieves superior performance over comparison algorithms.


Assuntos
Balistocardiografia/métodos , Frequência Cardíaca/fisiologia , Processamento de Sinais Assistido por Computador , Aprendizado de Máquina Supervisionado , Adulto , Algoritmos , Feminino , Humanos , Masculino , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...