Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nano Lett ; 19(5): 2968-2978, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30924343

RESUMO

Cisplatin-based chemotherapy is a widely used first-line strategy for numerous cancers. However, drug resistances are often inevitable accompanied by the long-term use of cisplatin in vivo, significantly hampering its therapeutic efficacy and clinical outcomes. Among others, autophagy induction is one of the most common causes of tumor resistance to cisplatin. Herein, a self-assembled nanoprodrug platform was developed with the synergistic effect of cisplatin and RNAi to fight against cisplatin-resistant lung cancer. The nanoprodrug platform consists of three molecular modules, including prodrug complex of Pt(IV)-peptide-bis(pyrene), DSPE-PEG, and cRGD-modified DSPE-PEG. The Pt(IV) is immobilized with peptide via amide bonds, allowing the Pt(IV) to be loaded with a loading efficiency of >95% and rapid-release active platinum ions (Pt(II)) in the presence of glutathione (GSH). Meanwhile, the peptide of the prodrug complex could efficiently deliver Beclin1 siRNA ( Beclin1 is an autophagy initiation factor) to the cytoplasm, thereby leading to autophagy inhibition. In addition, incorporation of DSPE-PEG and cRGD-modified DSPE-PEG molecules improves the biocompatibility and cellular uptake of the nanoprodrug platform. In vivo results also indicate that the nanoprodrug platform significantly inhibits the growth of a cisplatin-resistant tumor on xenograft mice models with a remarkable inhibition rate, up to 84% after intravenous injection.


Assuntos
Cisplatino/farmacologia , Neoplasias/tratamento farmacológico , Peptídeos/farmacologia , Pró-Fármacos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Proteína Beclina-1/química , Proteína Beclina-1/genética , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Linhagem Celular Tumoral , Cisplatino/efeitos adversos , Cisplatino/química , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Camundongos , Nanopartículas/química , Neoplasias/genética , Peptídeos/síntese química , Peptídeos/química , Pró-Fármacos/síntese química , Pró-Fármacos/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Small ; 13(33)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28677891

RESUMO

Autophagy is closely related to various diseases, and is a diagnostic and therapeutic target for some diseases. In recent years, tremendous efforts have been made to develop excellent probes for detection of autophagy. Nanostructure-based probes are interesting and promising approaches for in vivo biological imaging due to their unique structural and functional characteristics, e.g., modulating pharmacokinetics property by biocompatible coatings, multimodality capacity by delivering multiple imaging agents and highly specific targeting by antibody ligands. In this Review, we first introduce recent advancements in the development of nanostructure-based probes for detection of autophagy, including inorganic hybrid nanomaterials and self-assembled peptide polymeric nanoparticles. Meanwhile, a nanoprobe based on a "in vivo self-assembly" strategy is highlighted. The "in vivo self-assembly" endows nanoprobes with higher accumulation, and longer and better signal stability for in vivo detection of autophagy. Furthermore, this novel strategy could be widely used for biomedical imaging/diagnostics and therapeutics, which would attract more attention to this research area.


Assuntos
Autofagia , Nanotecnologia/métodos , Animais , Humanos , Sondas Moleculares/química , Nanoestruturas/química , Nanoestruturas/ultraestrutura
3.
Bioconjug Chem ; 28(6): 1709-1721, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28485595

RESUMO

Nanoscaled polymer-peptide conjugates (PPCs) containing both functional peptides and synthetic polymer comprise a new family of biomaterials that can circumvent the limitation of peptides alone. Our previous work showed that PPCs with the therapeutic peptide KLAK, especially PPCs with shorter PEG spacers and a higher degree of polymerization, exhibit enhanced antitumor effects through disrupting mitochondrial membranes. However, as PPCs have a spherical nanostructure (45-60 nm), this may have other effects besides the conjugated therapeutic peptide KLAK itself when they enter cancer cells. In this research, we compared the proteome differences of U87 cells treated with KLAK, polymer, and their conjugates (P-KLAK) through quantitative proteomics technology. The result reveals that proteins involved in oxidative stress response and the Nrf2/ARE pathway were significantly up-regulated after P-KLAK treatment. Moreover, the overexpression of sequestosome 1, a protein substrate that is selectively incorporated into the formation of autophagosome and degraded by autophagy, is found in our study and has not been reported previously in the study of KLAK toxicity. Additional experiments suggest that upon endocytosis, P-KLAK causes lysosome impairment and results in autophagosomes accumulation. Hence, P-KLAK might induce U87 cell death by autophagy blockage due to lysosome impairment as well as mitochondria damage synergistically.


Assuntos
Neoplasias/tratamento farmacológico , Peptídeos/química , Polímeros/química , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Lisossomos/efeitos dos fármacos , Mitocôndrias/patologia , Fator 2 Relacionado a NF-E2 , Neoplasias/patologia , Estresse Oxidativo , Peptídeos/uso terapêutico , Polímeros/uso terapêutico , Proteômica
4.
Small ; 12(21): 2921-31, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27120078

RESUMO

In drug delivery systems, pH-sensitive polymers are commonly used as drug carriers, and significant efforts have been devoted to the aspects of controlled delivery and release of drugs. However, few studies address the possible autophagic effects on cells. Here, for the first time, using a fluorescent autophagy-reporting cell line, this study evaluates the autophagy-induced capabilities of four types of pH-sensitive polymeric nanoparticles (NPs) with different physical properties, including size, surface modification, and pH-sensitivity. Based on experimental results, this study concludes that pH-sensitivity is one of the most important factors in autophagy induction. In addition, this study finds that variation of concentration of NPs could cause different autophagic effect, i.e., low concentration of NPs induces autophagy in an mTOR-dependent manner, but high dose of NPs leads to autophagic cell death. Identification of this tunable autophagic effect offers a novel strategy for enhancing therapeutic effect in cancer therapy through modulation of autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Lisossomos/química , Nanopartículas/química , Polímeros/química , Polímeros/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Células MCF-7
5.
Small ; 12(39): 5423-5430, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27511451

RESUMO

Au-BP7@SP nanohybrids with active motion under NIR laser irradiation can effectively enhance the temperature of tumor potentially by converting the kinetic energy to thermal energy, enhancing the killing efficiency of the tumor cells compared with Au@SP. The study provides an insight of nanohybrids' effect on photothermal treatment and opens a new avenue to cancer treatment by using self-propulsion Janus nanohybrids.


Assuntos
Hipertermia Induzida , Raios Infravermelhos , Neoplasias/terapia , Fototerapia , Animais , Sobrevivência Celular , Humanos , Células MCF-7 , Camundongos , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias/patologia , Temperatura
6.
Mol Pharm ; 12(8): 2869-78, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26101892

RESUMO

Various nanomaterials have been demonstrated as autophagy inducers owing to their endocytosis cell uptake pathway and impairment of lysosomes. pH-dependent nanomaterials as drug delivery systems that are capable of dissociating in weakly acidic lysosomal environment (pH 4-5) and consequently releasing the payloads into the cytoplasm have been paid extensive attention, but their autophagy-modulating effects are less reported so far. In this study, we report pH-sensitive micelle-like nanoparticles (NPs) that self-assembled from poly(ß-amino ester)s to induce cell autophagy. By encapsulation of gold(I) compounds (Au(I)) into hydrophobic domains of NPs, the resultant Au(I)-loaded NPs (Au(I)⊂NPs) shows synergistic cancer cell killing performance. The Au(I)⊂NPs enter cells through endocytosis pathway and accumulate into acidic lysosomes. Subsequently, the protonation of tertiary amines of poly(ß-amino ester)s triggers the dissociation of micelles, damages the lysosomes, and blocks formation of autolysosomes from fusion of lysosomes with autophagosomes. In addition, Au(I) preferentially inhibits thioredoxin reductase (TrxR) in MCF-7 human breast cancer cells that directly links to up-regulate reactive oxygen species (ROS) and consequently induce autophagy and apoptosis. The blockade of autophagy leads to excessive depletion of cellular organelles and essential proteins and ultimately results in cell death. Therefore, pH-sensitive polymeric nanoparticles with gold(I) compound payloads can synergistically induce cancer cell death through regulation of autophagy. Identification of the pH-sensitive nanomaterials for synergistically inducing cell death through regulation autophagy may open a new avenue for cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Ouro/química , Nanopartículas Metálicas/química , Polímeros/química , Linhagem Celular Tumoral , Portadores de Fármacos/química , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Lisossomos/metabolismo , Células MCF-7 , Micelas , Fagossomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-37096256

RESUMO

Owing to the breakthroughs in the prevention and control of the COVID-19 pandemic, messenger RNA (mRNA)-based vaccines have emerged as promising alternatives to conventional vaccine approaches for infectious disease prevention and anticancer treatments. Advantages of mRNA vaccines include flexibility in designing and manipulating antigens of interest, scalability in rapid response to new variants, ability to induce both humoral and cell-mediated immune responses, and ease of industrialization. This review article presents the latest advances and innovations in mRNA-based vaccines and their clinical translations in the prevention and treatment of infectious diseases or cancers. We also highlight various nanoparticle delivery platforms that contribute to their success in clinical translation. Current challenges related to mRNA immunogenicity, stability, and in vivo delivery and the strategies for addressing them are also discussed. Finally, we provide our perspectives on future considerations and opportunities for applying mRNA vaccines to fight against major infectious diseases and cancers. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.


Assuntos
Doenças Transmissíveis , Neoplasias , Vacinas , Humanos , Pandemias , RNA Mensageiro , Vacinas de mRNA , Neoplasias/prevenção & controle
8.
Adv Mater ; : e2306248, 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37897408

RESUMO

Smart nanorobots have emerged as novel drug delivery platforms in nanomedicine, potentially improving anti-cancer efficacy and reducing side effects. In this study, an intelligent tumor microenvironment-responsive nanorobot is developed that effectively delivers CpG payloads to Toll-like receptor 9 (TLR9)-positive tumors to induce autophagy-mediated cell death for immunotherapy. The nanorobots are fabricated by co-self-assembly of two amphiphilic triblock polymer peptides: one containing the matrix metallopeptidase 2 (MMP2)-cleaved GPLGVRGS motif to control the mechanical opening of the nanorobots and provide targeting capability for TLR-9-positive tumors and the other consisting of an arginine-rich GRRRDRGRS sequence that can condense nuclear acid payloads through electrostatic interactions. Using multiple tumor-bearing mouse models, it is investigated whether the intravenous injection of CpG-loaded nanorobots could effectively deliver CpG payloads to TLR-9-positive tumors and elicit anti-tumor immunity through TLR9 signaling and autophagy. Therefore, besides being a commonly used adjuvant for tumor vaccination, CpG-loaded nanorobots can effectively reprogram the tumor immunosuppressive microenvironment and suppress tumor growth and recurrence. This nanorobot-based CpG immunotherapy can be considered a feasible approach to induce anti-tumor immunity, showing great therapeutic potential for the future treatment of TLR9-positive cancers.

9.
Sci Transl Med ; 13(599)2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162754

RESUMO

Increasing clinical evidence has demonstrated that the deletion or mutation of tumor suppressor genes such as the gene-encoding phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in cancer cells may correlate with an immunosuppressive tumor microenvironment (TME) and poor response or resistance to immune checkpoint blockade (ICB) therapy. It is largely unknown whether the restoration of functional PTEN may modulate the TME and improve the tumor's sensitivity to ICB therapy. Here, we demonstrate that mRNA delivery by polymeric nanoparticles can effectively induce expression of PTEN in Pten-mutated melanoma cells and Pten-null prostate cancer cells, which in turn induces autophagy and triggers cell death-associated immune activation via release of damage-associated molecular patterns. In vivo results illustrated that PTEN mRNA nanoparticles can reverse the immunosuppressive TME by promoting CD8+ T cell infiltration of the tumor tissue, enhancing the expression of proinflammatory cytokines, such as interleukin-12, tumor necrosis factor-α, and interferon-γ, and reducing regulatory T cells and myeloid-derived suppressor cells. The combination of PTEN mRNA nanoparticles with an immune checkpoint inhibitor, anti-programmed death-1 antibody, results in a highly potent antitumor effect in a subcutaneous model of Pten-mutated melanoma and an orthotopic model of Pten-null prostate cancer. Moreover, the combinatorial treatment elicits immunological memory in the Pten-null prostate cancer model.


Assuntos
Melanoma/imunologia , Nanopartículas , PTEN Fosfo-Hidrolase , Neoplasias da Próstata/imunologia , Linhagem Celular Tumoral , Genes Supressores de Tumor , Humanos , Masculino , PTEN Fosfo-Hidrolase/genética , RNA Mensageiro/genética , Microambiente Tumoral
10.
Theranostics ; 10(1): 281-299, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31903120

RESUMO

RNA molecules (e.g., siRNA, microRNA, and mRNA) have shown tremendous potential for immunomodulation and cancer immunotherapy. They can activate both innate and adaptive immune system responses by silencing or upregulating immune-relevant genes. In addition, mRNA-based vaccines have recently been actively pursued and tested in cancer patients, as a form of treatment. Meanwhile, various nanomaterials have been developed to enhance RNA delivery to the tumor and immune cells. In this review article, we summarize recent advances in the development of RNA-based therapeutics and their applications in cancer immunotherapy. We also highlight the variety of nanoparticle platforms that have been used for RNA delivery to elicit anti-tumor immune responses. Finally, we provide our perspectives of potential challenges and opportunities of RNA-based nanotherapeutics in clinical translation towards cancer immunotherapy.


Assuntos
Vacinas Anticâncer/uso terapêutico , Portadores de Fármacos/química , Imunoterapia , Nanopartículas/química , Neoplasias/terapia , RNA/uso terapêutico , Humanos , Nanotecnologia
11.
Adv Biosyst ; 3(2): e1800232, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-32627370

RESUMO

Tumor-associated macrophages (TAMs) are of great interest in cancer immunology as they play an important role in the tumor microenvironment as cancer stromal cells recruited from circulating monocytes. TAMs are closely associated with tumor progression, including initiation, trophic growth, metabolism, angiogenesis, and metastasis; moreover, in clinical practice, their quantity can be related to poor prognosis. Fundamental and translational studies imply that TAMs are one of the most promising targets in tumor therapy. Herein, the biological origination and classification of TAMs, which correspond to their functions and differentiations, are reviewed in detail. In addition, recent basic research and clinical preprocess of TAMs in tumor immunotherapy are also discussed. Finally, the advances in the use of nanotechnology and TAMs for tumor therapy are discussed. This review focuses on the background and status of basic research and clinical significance of TAMs, points out the potential of TAMs in tumor immunological therapy, and clarifies the possibility of translation TAM-targeting therapies in medicine.


Assuntos
Imunoterapia , Macrófagos , Neoplasias , Microambiente Tumoral/imunologia , Animais , Humanos , Camundongos , Nanomedicina , Neoplasias/imunologia , Neoplasias/terapia
12.
J Med Chem ; 62(17): 7697-7707, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31381325

RESUMO

Aurora and polo-like kinases control the G2/M phase in cell mitosis, which are both considered as crucial targets for cancer cell proliferations. Here, naphthalene-based Aurora/PLK coinhibitors as leading compounds were designed through in silico approach, and a total of 36 derivatives were synthesized. One candidate (AAPK-25) was selected under in vitro cell based high throughput screening with an IC50 value = 0.4 µM to human colon cancer cell HCT-116. A kinome scan assay showed that AAPK-25 was remarkably selective to both Aurora and PLK families. The relevant genome pathways were also depicted by microarray based gene expression analysis. Furthermore, validated from a set of in vitro and in vivo studies, AAPK-25 significantly inhibited the development of the colon cancer growth and prolonged the median survival time at the end of the administration (p < 0.05). To sum up, AAPK-25 has a great potential to be developed for a chemotherapeutic agent in clinical use.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas , Inibidores de Proteínas Quinases/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Quinase 1 Polo-Like
13.
ACS Nano ; 13(7): 7568-7577, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31260255

RESUMO

Cellular immunotherapeutics aim to employ immune cells as anticancer agents. Ex vivo engineering of dendritic cells (DCs), the initial role of an immune response, benefits tumor elimination by boosting specific antitumor responses. However, directly activating DCs in vivo is less efficient and therefore quite challenging. Here, we designed a nanoactivator that manufactures DCs through autophagy upregulating in vivo directly, which lead to a high-efficiency antigen presention of DCs and antigen-specific T cells generation. The nanoactivator significantly enhances tumor antigen cross-presentation and subsequent T cell priming. Consequently, in vivo experiments show that the nanoactivators successfully reduce tumor growth and prolong murine survival. Taken together, these results indicate in situ DCs manipulation by autophagy induction is a promising strategy for antigen presentation enhancement and tumor elimination.


Assuntos
Autofagia/imunologia , Células Dendríticas/imunologia , Imunoterapia , Melanoma Experimental/terapia , Nanopartículas/química , Animais , Apresentação de Antígeno/imunologia , Linhagem Celular Tumoral , Feminino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Propriedades de Superfície , Linfócitos T/imunologia
14.
Nat Commun ; 10(1): 4861, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31649241

RESUMO

Achieving the activation of drugs within cellular systems may provide targeted therapies. Here we construct a tumour-selective cascade activatable self-detained system (TCASS) and incorporate imaging probes and therapeutics. We show in different mouse models that the TCASS system accumulates in solid tumours. The molecules show enhanced accumulation in tumour regions via the effect of recognition induced self-assembly. Analysis of the molecular penetration in tumour tissue shows that in vivo self-assembly increases the penetration capability compared to typical soft or hard nanomaterials. Importantly, the in vivo self-assembled molecules exhibit a comparable clearance pathway to that of small molecules, which are excreted from organs of the reticuloendothelial system (liver and kidney), while are relatively slowly eliminated from tumour tissues. Finally, this system, combined with the NIR probe, shows high specificity and sensitivity for detecting bladder cancer in isolated intact patient bladders.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Carcinoma de Células de Transição/diagnóstico por imagem , Corantes/administração & dosagem , Sistemas de Liberação de Medicamentos , Engenharia de Proteínas/métodos , Neoplasias da Bexiga Urinária/diagnóstico por imagem , Motivos de Aminoácidos , Animais , Disponibilidade Biológica , Carbocianinas/administração & dosagem , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Células HEK293 , Humanos , Rim/metabolismo , Fígado/metabolismo , Camundongos , Transplante de Neoplasias , Sensibilidade e Especificidade , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomaterials ; 156: 248-257, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29216535

RESUMO

The blockade of PD-1/PD-L1 interaction by peptide antagonists can unleash and enhance pre-existing anti-cancer immune responses of T cells to eradicate cancer cells. However, low proteolytic stability is the "Achilles' Heel" of peptides. Here, we first report a nanoantagonist with a physiological temperature sensitive nanophase-segregated surface that exhibits significantly enhanced blood circulation, peptide stability and PD-L1 immune checkpoint blockade efficacy. Thermosensitive polymers with different phase transition temperatures (Tt) are used to form the nanophase-segregated surface on an Au nanorod core. Importantly, the nanophase-segregated surface aids the nanoantagonist to resist protein adsorption and enhance the systemic stability of the linked peptides. Finally, the as-designed nanoantagonist effectively blocks PD-1/PD-L1 interaction in vitro and in vivo, enhances the pre-existing CD8+ T cell tumor destruction capability and inhibits tumor growth. This study offers a new strategy for designing nano-formulations for cancer immunotherapy.


Assuntos
Imunoterapia , Nanopartículas/química , Neoplasias/imunologia , Neoplasias/terapia , Adsorção , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antígeno B7-H1/metabolismo , Circulação Sanguínea/efeitos dos fármacos , Feminino , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Peptídeos/química , Polímeros/síntese química , Polímeros/química , Receptor de Morte Celular Programada 1/metabolismo , Ligação Proteica , Propriedades de Superfície , Temperatura , Distribuição Tecidual/efeitos dos fármacos
16.
Nat Commun ; 9(1): 1802, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29728565

RESUMO

Cerebral amyloid ß-peptide (Aß) accumulation resulting from an imbalance between Aß production and clearance is one of the most important causes in the formation of Alzheimer's disease (AD). In order to preserve the maintenance of Aß homeostasis and have a notable AD therapy, achieving a method to clear up Aß plaques becomes an emerging task. Herein, we describe a self-destructive nanosweeper based on multifunctional peptide-polymers that is capable of capturing and clearing Aß for the effective treatment of AD. The nanosweeper recognize and bind Aß via co-assembly through hydrogen bonding interactions. The Aß-loaded nanosweeper enters cells and upregulates autophagy thus promoting the degradation of Aß. As a result, the nanosweeper decreases the cytotoxicity of Aß and rescues memory deficits of AD transgenic mice. We believe that this resourceful and synergistic approach has valuable potential as an AD treatment strategy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Nanopartículas/química , Peptídeos/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Animais , Autofagia/efeitos dos fármacos , Proteína Beclina-1/química , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Linhagem Celular Tumoral , Quitosana/química , Modelos Animais de Doenças , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Peptídeos/administração & dosagem , Peptídeos/química , Placa Amiloide/química , Polietilenoglicóis/química
17.
ACS Nano ; 11(7): 7301-7311, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28628744

RESUMO

Intracellular construction of nanoaggregates from synthetic molecules to mimic natural ordered superstructures has gained increasing attention recently. Here, we develop an endogenous stimuli-induced aggregation (eSIA) approach to construct functional nanoaggregates for sensing and monitoring cellular physiological processes in situ. We design a series of thermosensitive polymer-peptide conjugates (PPCs), which are capable of constructing nanoaggregates in cells based on their isothermal phase transition property. The PPCs are composed of three moieties (i.e., a thermoresponsive polymer backbone, a grafted peptide, and a signal-molecule label). The bioenvironment-associated phase transition behavior of PPCs are carefully studied by consideration of various crucial parameters such as chain length, hydrophilicity, ratio of grafted peptides, and concentration. Intriguingly, under the specific intracellular stimulus, the PPCs are tailored and simultaneously form nanoaggregates exhibiting long-term retention effect, which enables specific identification and quantification of endogenous factors. This general approach is expected for high-performance in situ sensing and dynamic monitoring of disease progression in living subjects.


Assuntos
Nanoestruturas/química , Neoplasias/diagnóstico , Peptídeos/química , Transição de Fase , Polímeros/química , Animais , Antibióticos Antineoplásicos/uso terapêutico , Progressão da Doença , Doxorrubicina/uso terapêutico , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Confocal/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Neoplasias/terapia , Imagem Óptica/métodos , Prognóstico , Agregados Proteicos , Temperatura
18.
ACS Nano ; 11(4): 4086-4096, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28334523

RESUMO

Tumor metastasis is one of the big challenges in cancer treatment and is often associated with high patient mortality. Until now, there is an agreement that tumor invasion and metastasis are related to degradation of extracellular matrix (ECM) by enzymes. Inspired by the formation of natural ECM and the in situ self-assembly strategy developed in our group, herein, we in situ constructed an artificial extracellular matrix (AECM) based on transformable Laminin (LN)-mimic peptide 1 (BP-KLVFFK-GGDGR-YIGSR) for inhibition of tumor invasion and metastasis. The peptide 1 was composed of three modules including (i) the hydrophobic bis-pyrene (BP) unit for forming and tracing nanoparticles; (ii) the KLVFF peptide motif that was inclined to form and stabilize fibrous structures through intermolecular hydrogen bonds; and (iii) the Y-type RGD-YIGSR motif, derived from LN conserved sequence, served as ligands to bind cancer cell surfaces. The peptide 1 formed nanoparticles (1-NPs) by the rapid precipitation method, owing to strong hydrophobic interactions of BP. Upon intravenous injection, 1-NPs effectively accumulated in the tumor site due to the enhanced permeability and retention (EPR) effect and/or targeting capability of RGD-YIGSR. The accumulated 1-NPs simultaneously transformed into nanofibers (1-NFs) around the solid tumor and further entwined to form AECM upon binding to receptors on the tumor cell surfaces. The AECM stably existed in the primary tumor site over 72 h, which consequently resulted in efficiently inhibiting the lung metastasis in breast and melanoma tumor models. The inhibition rates in two tumor models were 82.3% and 50.0%, respectively. This in vivo self-assembly strategy could be widely utilized to design effective drug-free biomaterials for inhibiting the tumor invasion and metastasis.


Assuntos
Antineoplásicos/química , Matriz Extracelular/química , Neoplasias Pulmonares/terapia , Nanopartículas/química , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Humanos , Laminina/química , Neoplasias Pulmonares/patologia , Camundongos , Mimetismo Molecular , Nanofibras/química , Invasividade Neoplásica , Metástase Neoplásica , Tamanho da Partícula , Peptídeos/administração & dosagem , Peptídeos/química , Permeabilidade , Pirenos/química
19.
Biomaterials ; 141: 199-209, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28689116

RESUMO

Autophagic therapy is regarded as a promising strategy for disease treatment. Appropriate autophagy regulations in vivo play a crucial role in translating this new concept from benchside to bedside. So far, emerging technologies are required to spatially and quantitatively monitor autophagic process in vivo in order to minimize the cytotoxity concerns associated with autophagy-mediated therapy. We successfully demonstrate the "proof-of-concept" study on autophagy-mediated chemotherapy in mice. Here, we describe a photoacoustic (PA) nanoprobe based on "in vivo self-assembly" idea for real-time and quantitative detection of autophagy in mice for the first time. The purpurin-18 (P18) monomer is connected to hydrophilic poly(amidoamine) dendrimer (4th generation) through a peptide (GKGSFGFTG) that can be cleaved by an autophagy-specific enzyme, i.e., ATG4B, consequently resulting in aggregation of P18 and enhanced PA signals. Based on this aggregation-induced "turn-on" PA signals, we noninvasively determine the ATG4B activity for monitoring autophagy of tumor in vivo. According to the results of PA imaging, we could optimize chemotherapy efficacy through precisely modulating autophagy, which thereby decrease systemic toxicity from chemotherapeutics and autophagy inhibitors. We envision it will pave the way for developing autophagy-based treatment of diseases in the future.


Assuntos
Autofagia/efeitos dos fármacos , Técnicas Fotoacústicas/métodos , Porfirinas/metabolismo , Animais , Antineoplásicos/farmacologia , Proteínas Relacionadas à Autofagia/metabolismo , Cisteína Endopeptidases/metabolismo , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Poliaminas/química , Poliaminas/metabolismo , Porfirinas/química
20.
Biomaterials ; 112: 153-163, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27768970

RESUMO

Immunotherapy has shown promising treatment effects for a variety of cancers. However, the immune treatment efficiency for solid tumors is limited owing to insufficient infiltration of immune cells into solid tumors. The conversion of tumor-supportive macrophages to tumor-suppressive macrophages, inducing the functional reversal of macrophages, is an effective method and contributes to a subsequent antitumor response. The current challenge in the field is the poor distribution and systemic side effects associated with the use of cytokines. As a solution to this issue, we designed and synthesized microenvironment-responsive nanoparticles (P) with IL-12 payload (IL-12⊂P1). These nanoparticles could promote the systemic administration and release of IL-12 in the tumor microenvironment, and the locally responsive property of IL-12⊂P1 could subsequently re-educate tumor-associated macrophages (TAMs). In particular, our results illustrated the great therapeutic effects derived from the functional conversion of macrophages. Our strategy was to design a microenvironment-responsive material for local macrophage modification to overcome the physiological barrier of solid tumors. The shifting of macrophage phenotypes via IL-12⊂P1 achieved immunomodulation in the microenvironment for cancer therapy, with negligible cytotoxicity. We expect that the functional regulation of TAMs by pH-responsive nanomaterials is a promising therapeutic approach for cancer immunotherapy.


Assuntos
Interleucina-12/administração & dosagem , Macrófagos/imunologia , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/imunologia , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Feminino , Interleucina-12/química , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/patologia , Polímeros/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA