Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Proc Natl Acad Sci U S A ; 120(13): e2221874120, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36947515

RESUMO

Cyclic-di-GMP (c-di-GMP) is a ubiquitous bacterial signaling molecule. It is also a critical player in the regulation of cell size and cell behaviors such as cell aggregation and phototaxis in cyanobacteria, which constitute an important group of prokaryotes for their roles in the ecology and evolution of the Earth. However, c-di-GMP receptors have never been revealed in cyanobacteria. Here, we report the identification of a c-di-GMP receptor, CdgR, from the filamentous cyanobacterium Anabaena PCC 7120. Crystal structural analysis and genetic studies demonstrate that CdgR binds c-di-GMP at the dimer interface and this binding is required for the control of cell size in a c-di-GMP-dependent manner. Different functions of CdgR, in ligand binding and signal transmission, could be separated genetically, allowing us to dissect its molecular signaling functions. The presence of the apo-form of CdgR triggers cell size reduction, consistent with the similar effects observed with a decrease of c-di-GMP levels in cells. Furthermore, we found that CdgR exerts its function by interacting with a global transcription factor DevH, and this interaction was inhibited by c-di-GMP. The lethal effect triggered by conditional depletion of DevH or by the production of several point-mutant proteins of CdgR in cells indicates that this signaling pathway plays critical functions in Anabaena. Our studies revealed a mechanism of c-di-GMP signaling in the control of cell size, an important and complex trait for bacteria. CdgR is highly conserved in cyanobacteria, which will greatly expand our understanding of the roles of c-di-GMP signaling in these organisms.


Assuntos
Cianobactérias , Transdução de Sinais , Cianobactérias/metabolismo , GMP Cíclico/metabolismo , Regulação da Expressão Gênica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica
3.
Adv Mater ; 34(39): e2109701, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35906820

RESUMO

Polymer self-assembly is a crucial process in materials engineering. Currently, almost all polymer self-assembly is limited to non-covalent bonding methods, even though these methods have drawbacks as they require complicated synthesis techniques and produce relatively unstable structures. Here, a novel mechanism of covalent polymer self-assembly is discovered and employed to address drawbacks of non-covalent polymer self-assembly. A simple ketone homopolymer is found to self-assemble into nano- to macroscale hydrogels during covalent crosslinking. In contrast to non-covalent self-assembly, the covalent self-assembly is independent of and unaffected by solvent conditions (e.g., polarity and ionic strength) and does not require additional agents, e.g., organic solvents and surfactants. The covalent polymer self-assembly is subjected to a new mechanism of control by tuning the covalent crosslinking rate. This leads to nanogels with an unprecedented and tightly controlled range of dimensions from less than 10 nm to above 100 nm. Moreover, the crosslinking rate also regulates the assembly behavior of microgels fabricated by microfluidics. The microgels self-assemble into granular fibers, which is 3D printed into stable porous scaffolds. The novel covalent polymer assembly method has enormous potential to revolutionize multiscale materials fabrication for applications in drug delivery, tissue engineering, and many other fields.

4.
Theranostics ; 12(10): 4791-4801, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35832083

RESUMO

Background: Enzyme-activatable prodrugs are extensively employed in oncology and beyond. Because enzyme concentrations and their (sub)cellular compartmentalization are highly heterogeneous in different tumor types and patients, we propose ultrasound-directed enzyme-prodrug therapy (UDEPT) as a means to increase enzyme access and availability for prodrug activation locally. Methods: We synthesized ß-glucuronidase-sensitive self-immolative doxorubicin prodrugs with different spacer lengths between the active drug moiety and the capping group. We evaluated drug conversion, uptake and cytotoxicity in the presence and absence of the activating enzyme ß-glucuronidase. To trigger the cell release of ß-glucuronidase, we used high-intensity focused ultrasound to aid in the conversion of the prodrugs into their active counterparts. Results: More efficient enzymatic activation was observed for self-immolative prodrugs with more than one aromatic unit in the spacer. In the absence of ß-glucuronidase, the prodrugs showed significantly reduced cellular uptake and cytotoxicity compared to the parent drug. High-intensity focused ultrasound-induced mechanical destruction of cancer cells resulted in release of intact ß-glucuronidase, which activated the prodrugs, restored their cytotoxicity and induced immunogenic cell death. Conclusion: These findings shed new light on prodrug design and activation, and they contribute to novel UDEPT-based mechanochemical combination therapies for the treatment of cancer.


Assuntos
Neoplasias , Pró-Fármacos , Doxorrubicina/uso terapêutico , Glucuronidase/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico
5.
Artigo em Inglês | MEDLINE | ID: mdl-34637922

RESUMO

Toll like receptor 5 (TLR5) plays a crucial role in the innate immune response by recognizing bacterial flagellin proteins. In the present study, the genomic and 5'-flanking sequences of LcTLR5M (membrane) and LcTLR5S (soluble) were cloned from the large yellow croaker, Larimichthys crocea. Then, their promoter activities were determined in human embryonic kidney (HEK293T) cells. LcTLR5M contained 4 exons and 3 introns, and LcTLR5S contained 2 exons and 1 intron. Bioinformatic prediction of transcription factor binding sites (TFBSs) indicated that the promoter structures were different between LcTLR5M and LcTLR5S. A dual luciferase assay showed that the deletion mutant -471 to +189 of LcTLR5M promoter possessed the greatest activity with 36 times activity of the control (P < 0.05). For LcTLR5S, two deletion mutants, -1687 to +106 and - 720 to +106, showed high promoter activity. Furthermore, site-directed mutation demonstrated that a -392 to -391 AT/GG substitution in Oct-1 binding site, and a -104 to -103 GG/TT and a -98 to -97 CC/AA substitution in the NF-κB binding site of TLR5S caused a significant decline of luciferase activity (P < 0.05). Moreover, the co-transfection of an NF-κB/p65 over-expression plasmid with wild type TLR5S (-720 to +106) resulted in an extremely significant increase of promoter activity by more than 9 times compared with the NF-kB mutant (P < 0.01). Our findings suggest that the genomic organization and promoter structure may differ between LcTLR5M and LcTLR5S. Oct1 and NF-κB binding sites might be cis-regulatory elements in the LcTLR5S promoter. NF-κB/p65 plays an important role in LcTLR5S promoter activation through binding with the NF-κB binding site.


Assuntos
NF-kappa B , Perciformes , Animais , Sítios de Ligação , Proteínas de Peixes/genética , Células HEK293 , Humanos , Imunidade Inata , NF-kappa B/genética , NF-kappa B/metabolismo , Perciformes/genética , Perciformes/metabolismo
6.
Biomaterials ; 266: 120432, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33069116

RESUMO

Gastrointestinal (GI) cancers are among the most lethal malignancies. The treatment of advanced-stage GI cancer involves standard chemotherapeutic drugs, such as docetaxel, as well as targeted therapeutics and immunomodulatory agents, all of which are only moderately effective. We here show that Π electron-stabilized polymeric micelles based on PEG-b-p(HPMAm-Bz) can be loaded highly efficiently with docetaxel (loading capacity up to 23 wt%) and potentiate chemotherapy responses in multiple advanced-stage GI cancer mouse models. Complete cures and full tumor regression were achieved upon intravenously administering micellar docetaxel in subcutaneous gastric cancer cell line-derived xenografts (CDX), as well as in CDX models with intraperitoneal and lung metastases. Nanoformulated docetaxel also outperformed conventional docetaxel in a patient-derived xenograft (PDX) model, doubling the extent of tumor growth inhibition. Furthermore, micellar docetaxel modulated the tumor immune microenvironment in CDX and PDX tumors, increasing the ratio between M1-and M2-like macrophages, and toxicologically, it was found to be very well-tolerated. These findings demonstrate that Π electron-stabilized polymeric micelles loaded with docetaxel hold significant potential for the treatment of advanced-stage GI cancers.


Assuntos
Antineoplásicos , Neoplasias Gastrointestinais , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Docetaxel , Portadores de Fármacos , Elétrons , Neoplasias Gastrointestinais/tratamento farmacológico , Camundongos , Micelas , Polietilenoglicóis , Microambiente Tumoral
7.
J Control Release ; 328: 805-816, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33010332

RESUMO

Core-crosslinked polymeric micelles (CCPM) based on PEG-b-pHPMA-lactate are clinically evaluated for the treatment of cancer. We macroscopically and microscopically investigated the biodistribution and target site accumulation of CCPM. To this end, fluorophore-labeled CCPM were intravenously injected in mice bearing 4T1 triple-negative breast cancer (TNBC) tumors, and their localization at the whole-body, tissue and cellular level was analyzed using multimodal and multiscale optical imaging. At the organism level, we performed non-invasive 3D micro-computed tomography-fluorescence tomography (µCT-FLT) and 2D fluorescence reflectance imaging (FRI). At the tissue and cellular level, we performed extensive immunohistochemistry, focusing primarily on cancer, endothelial and phagocytic immune cells. The CCPM achieved highly efficient tumor targeting in the 4T1 TNBC mouse model (18.6 %ID/g), with values twice as high as those in liver and spleen (9.1 and 8.9 %ID/g, respectively). Microscopic analysis of tissue slices revealed that at 48 h post injection, 67% of intratumoral CCPM were localized extracellularly. Phenotypic analyses on the remaining 33% of intracellularly accumulated CCPM showed that predominantly F4/80+ phagocytes had taken up the nanocarrier formulation. Similar uptake patterns were observed for liver and spleen. The propensity of CCPM to primarily accumulate in the extracellular space in tumors suggests that the anticancer efficacy of the formulation mainly results from sustained release of the chemotherapeutic payload in the tumor microenvironment. In addition, their high uptake by phagocytic immune cells encourages potential use for immunomodulatory anticancer therapy. Altogether, the beneficial biodistribution, efficient tumor targeting and prominent engagement of PEG-b-pHPMA-lactate-based CCPM with key cell populations underline the clinical versatility of this clinical-stage nanocarrier formulation.


Assuntos
Micelas , Polímeros , Animais , Linhagem Celular Tumoral , Camundongos , Imagem Óptica , Distribuição Tecidual , Microtomografia por Raio-X
8.
Acta Pharmacol Sin ; 41(7): 954-958, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32555445

RESUMO

Cancer nanomedicines have shown promise in combination immunotherapy, thus far mostly preclinically but also already in clinical trials. Combining nanomedicines with immunotherapy aims to reinforce the cancer-immunity cycle, via potentiating key steps in the immune reaction cascade, namely antigen release, antigen processing, antigen presentation, and immune cell-mediated killing. Combination nano-immunotherapy can be realized via three targeting strategies, i.e., by targeting cancer cells, targeting the tumor immune microenvironment, and targeting the peripheral immune system. The clinical potential of nano-immunotherapy has recently been demonstrated in a phase III trial in which nano-albumin paclitaxel (Abraxane®) was combined with atezolizumab (Tecentriq®) for the treatment of patients suffering from advanced triple-negative breast cancer. In the present paper, besides strategies and initial (pre)clinical success stories, we also discuss several key challenges in nano-immunotherapy. Taken together, nanomedicines combined with immunotherapy are gaining significant attention, and it is anticipated that they will play an increasingly important role in clinical cancer therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoterapia , Nanomedicina , Neoplasias/terapia , Humanos , Neoplasias/imunologia , Neoplasias/patologia
9.
Chem Soc Rev ; 48(1): 351-381, 2019 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-30465669

RESUMO

Immunotherapy is revolutionizing the treatment of cancer. It can achieve unprecedented responses in advanced-stage patients, including complete cures and long-term survival. However, immunotherapy also has limitations, such as its relatively low response rates and the development of severe side effects. These drawbacks are gradually being overcome by improving our understanding of the immune system, as well as by establishing combination regimens in which immunotherapy is combined with other treatment modalities. In addition to this, in recent years, progress made in chemistry, nanotechnology and materials science has started to impact immuno-oncology, resulting in more effective and less toxic immunotherapy interventions. In this context, multiple different nanomedicine formulations and macroscale materials have been shown to be able to boost anti-cancer immunity and the efficacy of immunomodulatory drugs. We here review nanotechnological and materials chemistry efforts related to endogenous and exogenous vaccination, to the engineering of antigen-presenting cells and T cells, and to the modulation of the tumor microenvironment. We also discuss limitations, current trends and future directions. Together, the insights provided and the evidence obtained indicate that there is a bright future ahead for engineering nanomedicines and macroscale materials for immuno-oncology applications.


Assuntos
Imunoterapia , Substâncias Macromoleculares/química , Nanomedicina , Neoplasias/imunologia , Neoplasias/terapia , Humanos
10.
Fish Shellfish Immunol ; 74: 450-458, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29325713

RESUMO

Transforming growth factor-ß-activating kinase 1 (TAK1) is triggered by foreign pathogenic infection and involves in proinflammatory response through the activation of nuclear factor-κB (NF-κB), which is specifically regulated by TAK1-binding protein 1 (TAB1). However, the expression and regulatory characterizations of TAK1 and TAB1 in fish immune response remain largely unknown. In the present study, the cDNA sequences of TAK1 (LcTAK1) and TAB1 (LcTAB1) were identified from large yellow croaker, Larimichthys crocea. The open reading frame (ORF) of LcTAK1 was 1725 bp in length, encoding 574 amino acids. The putative LcTAK1 protein contained a protein kinase domain and a C-terminal coiled-coil region. The ORF of LcTAB1 was 1518 bp encoding 505 amino acids. And a typical PP2Cc domain and a conserved sequence motif (PYVDFSQFYLLWGSDH) at C-terminal were identified in the predicted LcTAB1 protein. Multiple alignments showed that LcTAK1 shared 74.0-97.9% and LcTAB1 shared 37.4-95.8% sequence identities with TAK1 and TAB1 proteins from other species, respectively. Quantitative PCR analysis indicated that both LcTAK1 and LcTAB1 were broadly expressed in all examined tissues, with the most predominant expression in brain and the weakest expression in muscle, respectively. Subcellular localization revealed that both LcTAK1 and LcTAB1 expressed in the cytoplasm. In addition, LcTAK1 transcripts increased significantly in LCK cells after flagellin, LPS and poly I:C stimulation while LcTAB1 enhanced greatly after LPS and poly I:C challenge. Furthermore, the roles of them in NF-κB activation were investigated by overexpression of LcTAK1 and LcTAB1 in HEK293T cells. Our results revealed that NF-κB luciferase promoter expression could not be induced by overexpression of LcTAK1 or LcTAB1 alone, however, it could be induced by co-expression of LcTAK1 and LcTAB1 together. Moreover, the roles of LcTAK1 and LcTAB1 in immune response analysis showed that NF-κB activation enhanced significantly in co-overexpressed HEK293T cells following LPS and poly I:C stimulation. However, the expression levels of tumor necrosis factor (TNF)-α, Interleukin-6 (IL-6) and IL-8 were induced only after LPS challenge (p < .05). These findings suggested that the TAK1-TAB1 complex of large yellow croaker might play an important role in pro-inflammatory cytokines and chemokine release after LPS stimulation via inducing NF-κB activation.


Assuntos
Citocinas/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Regulação da Expressão Gênica , Imunidade Inata/genética , Perciformes/genética , Perciformes/imunologia , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Peixes/química , Perfilação da Expressão Gênica/veterinária , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Filogenia
11.
Biomacromolecules ; 18(5): 1449-1459, 2017 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-28328191

RESUMO

Tumor-targeted nanomedicines have been extensively applied to alter the drawbacks and enhance the efficacy of chemotherapeutics. Despite the large number of preclinical nanomedicine studies showing initial success, their therapeutic benefit in the clinic has been rather modest, which is partially due to the inefficient tumor penetration caused by the tumor microenvironment (high density of cells and extracellular matrix, increased interstitial fluid pressure). Furthermore, tumor penetration of nanomedicines is significantly influenced by physicochemical characteristics, such as size, surface chemistry, and shape. The effect of size on tumor penetration has been exploited to design nanomedicines with switchable size to tackle this challenge. Moreover, several pharmacological and physical approaches have been developed to enhance the tumor penetration of nanomedicines, by penetration-promoting ligands, intratumoral drug release, and modulating the tumor microenvironment and vasculature. Overall, these efforts have resulted in nanomedicines with better tumor penetration properties and with enhanced therapeutic efficacy. Future research should be directed to penetration-promoting strategies with broad applicability and with high translational potential.


Assuntos
Nanomedicina/métodos , Nanopartículas/metabolismo , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Humanos , Microambiente Tumoral
12.
J Drug Target ; 19(1): 49-55, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20205530

RESUMO

The aim of this study was to evaluate the pharmacokinetics (PK), tissue distribution, and the specific drug targeting of cefpiramide sodium-loaded liposomes (CPMS-Lips) compared with cefpiramide sodium solution (CPMS-Sol) in mice. CPMS-Lips were prepared by reverse phase evaporation method. In the PK and biodistribution study, mice received a single intravenous (i.v.) injection of 152 mg/kg of either CPMS-Lips or CPMS-Sol. Plasma and tissues were treated using liquid-liquid extraction and determined using reversed-phase high-performance liquid chromatography (RP-HPLC). The results showed that the CPMS-Lips prepared in this study had an average diameter of 7.146 ± 0.29 µm. In the plasma, the bioavailability (F) and the mean residence times (MRT) of the CPMS-Lips were 2.8- and 4.5-fold larger, respectively, than those of CPMS-Sol. CPMS-Lips also showed a significant difference in the tissue distribution profile in mice when compared with the conventional. The value of the intake rate (r(e)) for the lung was 2.97, which was the highest among the tested tissues. Meanwhile, the ratio of targeting efficiency (Te(liposome)/Te(injection)) of lung to that of other tissues for CPMS-Lips elevated significantly. These showed that CPMS-Lips can improve the bioavailability and biodistribution of CPMS in the lung. In conclusion, the liposome was a promising sustained-release and drug-targeting system for antibiotic drugs.


Assuntos
Antibacterianos/farmacocinética , Cefalosporinas/farmacocinética , Sistemas de Liberação de Medicamentos , Pulmão/metabolismo , Animais , Antibacterianos/administração & dosagem , Disponibilidade Biológica , Cefalosporinas/administração & dosagem , Cromatografia Líquida de Alta Pressão , Preparações de Ação Retardada , Feminino , Injeções Intravenosas , Lipossomos , Masculino , Camundongos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA