Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Chem Rev ; 123(12): 7782-7853, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37186942

RESUMO

The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.


Assuntos
Anticorpos , Imunoconjugados , Anticorpos/uso terapêutico , Imunoconjugados/uso terapêutico , Biotecnologia , Oligonucleotídeos
2.
J Am Chem Soc ; 145(44): 24272-24283, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37899626

RESUMO

Targeted degradation of the cell-surface and extracellular proteins via the endogenous lysosomal degradation pathways, such as lysosome-targeting chimeras (LYTACs), has recently emerged as an attractive tool to expand the scope of extracellular chemical biology. Herein, we report a series of recombinant proteins genetically fused to insulin-like growth factor 2 (IGF2), which we termed iLYTACs, that can be conveniently obtained in high yield by standard cloning and bacterial expression in a matter of days. We showed that both type-I iLYTACs, in which IGF2 was fused to a suitable affibody or nanobody capable of binding to a specific protein target, and type-II iLYTAC (or IGF2-Z), in which IGF2 was fused to the IgG-binding Z domain that served as a universal antibody-binding adaptor, could be used for effective lysosomal targeting and degradation of various extracellular and membrane-bound proteins-of-interest. These heterobifunctional iLYTACs are fully genetically encoded and can be produced on a large scale from conventional E. coli expression systems without any form of chemical modification. In the current study, we showed that iLYTACs successfully facilitated the cell uptake, lysosomal localization, and efficient lysosomal degradation of various disease-relevant protein targets from different mammalian cell lines, including EGFR, PD-L1, CD20, and α-synuclein. The antitumor properties of iLYTACs were further validated in a mouse xenograft model. Overall, iLYTACs represent a general and modular strategy for convenient and selective targeted protein degradation, thus expanding the potential applications of current LYTACs and related techniques.


Assuntos
Escherichia coli , Proteínas de Membrana , Humanos , Camundongos , Animais , Proteínas de Membrana/metabolismo , Escherichia coli/metabolismo , Transdução de Sinais , Lisossomos/metabolismo , Linhagem Celular , Mamíferos/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia
3.
Acc Chem Res ; 55(24): 3663-3674, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36484537

RESUMO

Although lipids are not genetically encoded, they are fundamental building blocks of cell membranes and essential components of cell metabolites. Lipids regulate various biological processes, including energy storage, membrane trafficking, signal transduction, and protein secretion; therefore, their metabolic imbalances cause many diseases. Approximately 47 000 lipid species with diverse structures have been identified, but little is known about their crucial roles in cellular systems. Particularly the structural, metabolic, and signaling functions of lipids often arise from interactions with proteins. Lipids attach to proteins not only by covalent bonds but also through noncovalent interactions, which also influence protein functions and localization. Therefore, it is important to explore this lipid-protein "interactome" to understand its roles in health and disease, which may further provide insight for medicinal development. However, lipid structures are generally quite complicated, rendering the systematic characterization of lipid-protein interactions much more challenging.Chemoproteomics is a well-known chemical biology platform in which small-molecule chemical probes are utilized in combination with high-resolution, quantitative mass spectrometry to study protein-ligand interactions in living cells or organisms, and it has recently been applied to the study of protein-lipid interactions as well. The study of these complicated interactions has been advanced by the development of bifunctional lipid probes, which not only enable probes to form covalent cross-links with lipid-interacting proteins under UV irradiation, but are also capable of enriching these proteins through bioorthogonal reactions.In this Account, we will discuss recent developments in bifunctional lipid-derived, affinity-based probes (AfBP)s that have been developed to investigate lipid-protein interactions in live cell systems. First, we will give a brief introduction of fundamental techniques based on AfBPs which are related to lipid research. Then, we will focus on three aspects, including probes developed on the basis of lipidation, lipid-derived probes with different modification positions (e.g., hydrophobic or hydrophilic parts of a lipid), and, finally, in situ biosynthesis of probes through intrinsic metabolic pathways by using chemically modified building blocks. We will present some case studies to describe these probes' design principles and cellular applications. At the end, we will also highlight key limitations of current approaches so as to provide inspirations for future improvement. The lipid probes that have been constructed are only the tip of the iceberg, and there are still plenty of lipid species that have yet to be explored. We anticipate that AfBP-based chemoproteomics and its further advancement will pave the way for a deep understanding of lipid-protein interactions in the future.


Assuntos
Proteínas , Transdução de Sinais , Proteínas/química , Membrana Celular/metabolismo , Lipídeos/química
4.
Angew Chem Int Ed Engl ; 61(21): e202200303, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35302274

RESUMO

Lysine acylation plays pivotal roles in cell physiology, including DNA transcription and repair, signal transduction, immune defense, metabolism, and many other key cellular processes. Molecular mechanisms of dysregulated lysine acylation are closely involved in the pathophysiological progress of many human diseases, most notably cancers. In recent years, chemical biology tools have become instrumental in studying the function of post-translational modifications (PTMs), identifying new "writers", "erasers" and "readers", and in targeted therapies. Here, we describe key developments in chemical biology approaches that have advanced the study of lysine acylation and its regulatory proteins (2016-2021). We further discuss the discovery of ligands (inhibitors and PROTACs) that are capable of targeting regulators of lysine acylation. Next, we discuss some current challenges of these chemical biology probes and suggest how chemists and biologists can utilize chemical probes with more discriminating capacity. Finally, we suggest some critical considerations in future studies of PTMs from our perspective.


Assuntos
Lisina Acetiltransferases , Lisina , Acilação , Biologia , Humanos , Lisina/metabolismo , Lisina Acetiltransferases/metabolismo , Processamento de Proteína Pós-Traducional
5.
Chemistry ; 27(47): 12207-12214, 2021 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-34115398

RESUMO

Mitochondria are key organelles that perform vital cellular functions such as those related to cell survival and death. The targeted delivery of different types of cargos to mitochondria is a well-established strategy to study mitochondrial biology and diseases. Of the various existing mitochondrion-transporting vehicles, most suffer from poor cytosolic entry, low delivery efficiency, limited cargo types, and cumbersome preparation protocols, and none was known to be universally applicable for mitochondrial delivery of different types of cargos (small molecules, proteins, and nanomaterials). Herein, two new cell-penetrating, mitochondrion-targeting ligands (named MitoLigand ) that are capable of effectively "tagging" small-molecule drugs, native proteins and nanomaterials are disclosed, as well as their corresponding chemoselective conjugation chemistry. Upon successful cellular delivery and rapid endosome escape, the released native cargos were found to be predominantly localized inside mitochondria. Finally, by successfully delivering doxorubicin, a well-known anticancer drug, to the mitochondria of HeLa cells, we showed that the released drug possessed potent cell cytotoxicity, disrupted the mitochondrial membrane potential and finally led to apoptosis. Our strategy thus paves the way for future mitochondrion-targeted therapy with a variety of biologically active agents.


Assuntos
Peptídeos Penetradores de Células , Nanoestruturas , Sistemas de Liberação de Medicamentos , Células HeLa , Humanos , Ligantes , Mitocôndrias
6.
J Am Chem Soc ; 142(1): 417-423, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31846316

RESUMO

Sensors capable of detecting different types of biomolecules have widespread applications in the field of biomedical research, but despite many years of research, the development of biosensors suitable for point-of-care (POC) applications in resource-limited areas is still extremely challenging. Sensors based on photonic crystal hydrogels (PCHs) hold much promise in this regard because of their numerous advantages over other existing bioanalytical methods. All current PCH biosensors are however restricted in the types of analytes they can detect sensitively with good selectivity. By taking advantage of the powerful and ubiquitous antibody-antigen interaction, we report herein the first-ever competition-based PCH biosensors capable of naked-eye detection of various biomolecules (e.g., proteins, peptides, and small molecules) with high sensitivity and selectivity and minimal background and excellent reversibility. We showed such PCH designs could be extended to the fabrication of different enzyme-detecting biosensors. The universal feature of these novel biosensors thus enables future development of POC biosensors in disease diagnostics for other bioanalytes.


Assuntos
Técnicas Biossensoriais , Fótons , Polímeros/química , Semicondutores , Energia Solar , Cristalização , Imidas/química , Luminescência , Naftalenos/química , Espalhamento de Radiação , Espectrofotometria Ultravioleta
7.
J Am Chem Soc ; 140(47): 15986-15996, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30384589

RESUMO

Therapeutic proteins have increased dramatically in both number and frequency of use in recent years, primarily owing to advances in protein engineering. Protein therapy provides the advantages of high potency and specificity, as well as low oncogenic risks. To date, due to their inability to cross the plasma membrane into the intracellular space of mammalian cells, most therapeutic proteins can only target secreted modulators or extracellular receptors. The full potential of protein therapy is, however, being gradually realized by the development of various strategies capable of intracellular protein delivery. Notwithstanding, most of these strategies suffer from severe endosomal trapping, resulting in very low protein delivery efficiency. In this Perspective, we discuss various methods to directly transport proteins into the cell cytoplasm, thus bypassing the problems associated with endocytosis.


Assuntos
Proteínas/farmacocinética , Transporte Biológico , Membrana Celular/metabolismo , Citosol/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Endocitose , Células HeLa , Humanos , Proteínas/metabolismo , Proteínas/farmacologia
8.
Chembiochem ; 19(9): 986-996, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29465822

RESUMO

Standard small-molecule microarrays (SMMs) are not well-suited for cell-based screening assays. Of the few attempts made thus far to render SMMs cell-compatible, all encountered major limitations. Here we report the first mesoporous silica nanoparticle (MSN)-on-a-chip platform capable of allowing high-throughput cell-based screening to be conducted on SMMs. By making use of a glass surface on which hundreds of MSNs, each encapsulated with a different native natural product, were immobilized in spatially defined manner, followed by on-chip mammalian cell growth and on-demand compound release, high-content screening was successfully carried out with readily available phenotypic detection methods. By combining this new MSN-on-a-chip system with small interfering RNA technology for the first time, we discovered that (+)-usniacin possesses synergistic inhibitory properties similar to those of olaparib (an FDA-approved drug) in BRCA1-knockdown cancer cells.


Assuntos
Produtos Biológicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Dispositivos Lab-On-A-Chip , Nanopartículas/química , Dióxido de Silício/química , Células A549 , Avaliação Pré-Clínica de Medicamentos/métodos , Desenho de Equipamento , Células HeLa , Ensaios de Triagem em Larga Escala/métodos , Humanos , Nanopartículas/ultraestrutura , Porosidade
9.
Angew Chem Int Ed Engl ; 57(6): 1532-1536, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29251821

RESUMO

Intracellular delivery of therapeutic proteins is highly challenging and in most cases requires chemical or genetic modifications. Herein, two complementary approaches for endocytosis-independent delivery of proteins to live mammalian cells are reported. By using either a "glycan" tag naturally derived from glycosylated proteins or a "traceless" tag that could reversibly label native lysines on non-glycosylated proteins, followed by bioorthogonal conjugation with cell-penetrating poly(disulfide)s (CPDs), we achieved intracellular delivery of proteins (including antibodies and enzymes) which, upon spontaneous degradation of CPDs, led to successful release of their "native" functional forms with immediate bioavailability.


Assuntos
Dissulfetos/química , Proteínas/metabolismo , Transfecção/métodos , Anticorpos/química , Anticorpos/metabolismo , Ciclo-Octanos/química , Endocitose , Glutationa/química , Células HeLa , Peroxidase do Rábano Silvestre/química , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Nanocápsulas/química , Processamento de Proteína Pós-Traducional , Proteínas/química , Ribonuclease Pancreático/química , Ribonuclease Pancreático/metabolismo
10.
Angew Chem Int Ed Engl ; 56(41): 12481-12485, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28816384

RESUMO

Antibodies are important biopharmaceuticals, but almost all existing antibody-based drugs are limited to targeting antigens located at the cell exterior because of the inability of antibodies to enter the cell interior. Available methods for intracellular delivery of antibodies have major shortcomings. Herein, we report an approach to encapsulate native antibodies in a biodegradable silica nanoquencher (BS-qNP), which could undergo efficient cellular uptake and intracellular degradation to release antibodies only under hypoxic conditions. By coating the surface of BS-qNP with cell-penetrating poly(disulfide)s (CPD), the delivered antibodies (or other proteins) avoided endolysosomal trapping. Doping of the silica coating with a fluorescent dye and a dark hole quencher further endowed BS-qNP with hypoxia-responsive fluorescence turn-on property. Our antibody delivery system thus provides the first platform capable of stable encapsulation, efficient uptake, on-demand antibody release, and imaging of release/cell state.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Cetuximab/administração & dosagem , Preparações de Ação Retardada/química , Nanopartículas/química , Dióxido de Silício/química , Células A549 , Animais , Antineoplásicos Imunológicos/farmacocinética , Células CHO , Hipóxia Celular , Cetuximab/farmacocinética , Cricetulus , Dissulfetos/química , Humanos , Nanopartículas/ultraestrutura
11.
Angew Chem Int Ed Engl ; 56(21): 5829-5833, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28429463

RESUMO

The protein-lipid interaction is an essential metabolic process that mediates cellular signaling and functions. Existing strategies for large-scale mapping studies of the protein-lipid interaction fall short in their incompatibility with metabolic incorporation or inability to remove unwanted interferences from lipidated proteins. By incorporating an alkyne-containing choline head group and a diazirine-modified fatty acid simultaneously into choline-containing phospholipids synthesized from live mammalian cells, protein-phospholipid interactions have been successfully imaged in live cells. Subsequent in situ profiling of the modified Cho phospholipid-crosslinked proteins followed by quantitative proteomics allowed identification of several hundred putative phospholipid-interacting proteins, some of which were further validated.


Assuntos
Colina/química , Lipídeos/química , Fosfolipídeos/química , Proteínas/química , Proteômica , Células Cultivadas , Cromatografia Líquida , Humanos , Espectrometria de Massas
12.
Angew Chem Int Ed Engl ; 55(16): 4933-7, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-26971527

RESUMO

Newly synthesized proteins constitute an important subset of the proteome involved in every cellular process, yet existing chemical tools used to study them have major shortcomings. Herein we report a suite of cell-permeable puromycin analogues capable of being metabolically incorporated into newly synthesized proteins in different mammalian cells, including neuronal cells. Subsequent labeling with suitable bioorthogonal reporters, in both fixed and live cells, enabled direct imaging and enrichment of these proteins. By taking advantage of the mutually orthogonal reactivity of these analogues, we showed multiplexed labeling of different protein populations, as well as quantitative measurements of protein dynamics by fluorescence correlation spectroscopy, could be achieved in live-cell environments.


Assuntos
Neurônios/citologia , Biossíntese de Proteínas , Puromicina/química , Células HeLa , Humanos
13.
Angew Chem Int Ed Engl ; 54(32): 9390-3, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26179829

RESUMO

A pentanidium-catalyzed highly enantioselective conjugate addition of 3-alkyloxindoles to phenyl vinyl sulfone has been demonstrated. This approach allows the construction of 3,3-dialkyl-substituted oxindole frameworks with high yield and excellent enantioselectivity (up to 99%) under simple phase-transfer conditions. A variety of oxindoles bearing all-carbon quaternary stereogenic centers were obtained in the presence of 0.25 mol% pentanidium. Meanwhile, practicality was illustrated by a gram-scale asymmetric synthesis of two 3,3-dialkyl-substituted oxindoles. The resulting adduct can be smoothly transformed to the natural product analogue in a short synthetic route.

14.
ACS Cent Sci ; 9(10): 1864-1893, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37901179

RESUMO

Cancer immunotherapy has become an established therapeutic paradigm in oncologic therapy, but its therapeutic efficacy remains unsatisfactory in the majority of cancer patients. Accumulating evidence demonstrates that the metabolically hostile tumor microenvironment (TME), characterized by acidity, deprivation of oxygen and nutrients, and accumulation of immunosuppressive metabolites, promotes the dysfunction of tumor-infiltrating immune cells (TIICs) and thereby compromises the effectiveness of immunotherapy. This indicates the potential role of tumor metabolic intervention in the reinvigoration of antitumor immunity. With the merits of multiple drug codelivery, cell and organelle-specific targeting, controlled drug release, and multimodal therapy, tumor metabolism-rewriting nanomedicines have recently emerged as an attractive strategy to strengthen antitumor immune responses. This review summarizes the current progress in the development of multifunctional tumor metabolism-rewriting nanomedicines for evoking antitumor immunity. A special focus is placed on how these nanomedicines reinvigorate innate or adaptive antitumor immunity by regulating glucose metabolism, amino acid metabolism, lipid metabolism, and nucleotide metabolism at the tumor site. Finally, the prospects and challenges in this emerging field are discussed.

15.
Biomaterials ; 294: 122000, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36640541

RESUMO

Cell nucleus is the desired subcellular organelle of many therapeutic drugs. Although numerous nanomaterial-based methods have been developed which could facilitate nuclear-targeted delivery of small-molecule drugs, few are known to be capable of delivering exogenous native proteins. Herein, we report a convenient and highly robust approach for effective nuclear-targeted delivery of native proteins/antibodies by using biodegradable silica nanocapsules (BSNPs) that were surface-modified with different nuclear localization signals (NLS) peptides. We found that, upon gaining entry to mammalian cells via endocytosis, such nanocapsules (protein@BSNP-NLS) could effectively escape from endolysosomal vesicles with the assistance of an endosomolytic peptide (i.e., L17E), accumulate in cell nuclei and release the encapsulated protein cargo with biological activities. Cloaked with HeLa cell membrane, DNase@BSNP-NLS/L17E-M (with L17E encapsulated) homologously delivered functional proteins to cancer cell nuclei in tumor-xenografted mice. In vitro and in vivo anti-tumor properties, such as long blood circulation time and effective tumor growth inhibition, indicate that the nuclear-targeted cell-membrane-cloaked BSNPs (DNase@BSNP-NLS/L17E-M) platform is a promising therapeutic approach to nuclear related diseases.


Assuntos
Nanocápsulas , Neoplasias , Humanos , Animais , Camundongos , Nanocápsulas/química , Células HeLa , Proteínas/metabolismo , Peptídeos/química , Sinais de Localização Nuclear , Desoxirribonucleases/metabolismo , Núcleo Celular/metabolismo , Mamíferos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
16.
Biomaterials ; 281: 121376, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35065331

RESUMO

Combination therapy is a promising strategy for treating multidrug-resistant (MDR) cancers. Macromolecules such as antibodies and RNAs have been successfully used for targeted therapy owing to their high specificity. However, their application as therapeutics remains limited due to membrane impermeability and poor intracellular stability. Designing drug delivery systems capable of co-administering macromolecules is therefore crucial for advancing them as therapeutics for combination therapy. Herein, by using glutathione (GSH)-responsive biodegradable silica nanocapsules (BS-NPs), we report for the first time a highly versatile nanomaterial-based strategy for co-encapsulation and intracellular co-delivery of different combinations of macromolecules (i.e., siRNA/protein, siRNA/antibody and protein/antibody). This strategy was successfully used in the intracellular co-delivery of siRNA/Cetuximab (also named Erbitux™) for combination therapy in epidermal growth factor receptor (EGFR)-overexpressing cancer cells. These BS-NPs showed good biosafety profiles and antitumor efficacy when administered in vivo, suggesting that the strategy holds potential as a novel delivery platform for combination cancer therapy.


Assuntos
Nanocápsulas , Nanopartículas , Cetuximab/uso terapêutico , Sistemas de Liberação de Medicamentos , Glutationa , RNA Interferente Pequeno/genética , Dióxido de Silício
17.
Chem Commun (Camb) ; 58(11): 1792-1795, 2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35040443

RESUMO

Monitoring gene delivery has significant benefits in gene therapy. Herein, we report a nanoquencher system by doping a FRET pair during nucleic acid-assisted cell penetrating poly(disulfide) (CPD) formation. Our results show that this strategy not only produces an efficient gene delivery polymer with minimal endolysosomal trapping, but also enables monitoring the release of the gene from the vehicle in live cells. This study further expanded the application of CPDs as promising tools in gene delivery.


Assuntos
Dissulfetos
18.
ACS Chem Biol ; 17(1): 32-38, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-34936351

RESUMO

Protein therapy provides a powerful alternative to small-molecule-based therapy, especially on cellular targets that are normally considered to be less druggable. Intracellular protein delivery, in particular, in a cell-type-specific manner, is still highly challenging. At present, few general strategies are available for the robust and selective intracellular delivery of proteins. In this Letter, by using zeolitic imidazolate framework-8 (ZIF-8) as protein-encapsulated nanoparticles and simultaneous doping with norbornene-modified imidazole (MIM-Nor), followed by surface attachment of the resulting nanoparticles with cetuximab (Cet) through click chemistry, we successfully synthesized Cet@protein@ZIF-8N, which was subsequently used for the selective intracellular delivery of functional proteins to epidermal-growth-factor-receptor (EGFR)-overexpressed cells. Both in-cell and in vivo experiments proved that Cet@RNase A@ZIF-8N can effectively deliver RNase A with the retention of selective inhibition. Furthermore, the same strategy was successfully applied to cell-type-specific gene editing through the delivery of a Cas9/sgRNA complex to knockdown the endogenous expression of glutathione peroxidase (GPX4), a key protein in ferroptosis. Our new system thus has potential implications in future cancer treatment and the development of precision medicine.


Assuntos
Química Click , Edição de Genes/métodos , Imidazóis/química , Estruturas Metalorgânicas/química , Neoplasias/metabolismo , Linhagem Celular Tumoral , Receptores ErbB , Técnicas de Transferência de Genes , Humanos , Nanopartículas/química
19.
ACS Sens ; 6(8): 3102-3111, 2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34383471

RESUMO

Colloidal inorganic nanostructures (metal, carbon, and silica) have been widely used as "nanoquenchers" for construction of nanosensors; however, inherent drawbacks such as insufficient fluorescence quenching efficiency, false positive signals, and uncertain long-term cytotoxicity have limited their further utility. Herein, by taking advantages of polymeric nanoparticles (PNPs) in terms of high loading capacity, facile surface modification chemistry, and good biocompatibility, we report a broad-spectrum (400-750 nm) polymeric fluorescence-quenching platform for sensor fabrication. Our newly developed polymeric nanoquenchers (qPNPs) were constructed by concurrently encapsulating various alkylated black-hole quenchers into nanoparticles made of poly(methyl methacrylate-co-methacrylic acid) and were found to have an excellent fluorescence quenching effect (>400-fold) on common fluorophores (FAM, TMR, and Cy5) together with high stability under physiological conditions. As a proof of concept, the feasibility of these qPNPs for fluorescence sensing was validated by successful construction of two nanosensors (FAMDEVD@qPNP and Cy5SurC@qPNP), which could be used as promising nanosensors for live-cell imaging of the apoptosis-related protease caspase-3 and cancer-related survivin mRNA, respectively. As expected, in the FAM channel, the FAMDEVD@qPNP showed fast and selective fluorescence responses toward caspase-3 in buffers and could be used to image the activation of drug-induced endogenous caspase-3. In the Cy5 channel, the Cy5SurC@qPNP could be used to distinguish normal cells (MCF10A) from cancer cells (HeLa) by quantitatively detecting the endogenous survivin mRNA level. It could be further used to monitor changes in the endogenous survivin mRNA expression levels in drug-treated HeLa cells. Altogether, by virtue of their high quencher loading and broad-spectrum quenching efficiency and good signal-to-background ratio, these qPNPs might be particularly attractive alternatives to other conventional nanoquenchers for the construction of more complex biosensors in the future.


Assuntos
Técnicas Biossensoriais , Nanoestruturas , Corantes Fluorescentes , Células HeLa , Humanos , Polímeros
20.
J Pharm Anal ; 10(5): 414-425, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33133725

RESUMO

Tyrosinase is an important enzyme in controlling the formation of melanin in melanosome, and plays a key role in the pigmentation of hair and skin. The abnormal expression or activation of tyrosinase is associated with several diseases such as albinism, vitiligo, melanoma and Parkinson disease. Excessive deposition of melanin could cause diseases such as freckles and brown spots in the human body, and it is also closely related to browning of fruits and vegetables and insect molting. Detecting and inhibiting the activity of tyrosinase is of extraordinary value in the progress of diagnosis and treatment of these diseases. Therefore, many selective optical detection probes and small molecular inhibitors have been developed, and have made significant contributions to the basic and clinical research on these diseases. In this paper, the detection and inhibition of tyrosinase and their application in whitening products are reviewed, with special emphasis on development of fluorescent probes and inhibitors. Hopefully, this review will help design more efficient and sensitive tyrosinase probes and inhibitors, as well as shed light on novel treatment of diseases such as melanoma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA