Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Am Chem Soc ; 146(1): 330-341, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38113388

RESUMO

Implementing dissipative assembly in living systems is meaningful for creation of living materials or even artificial life. However, intracellular dissipative assembly remains scarce and is significantly impeded by the challenges lying in precisely operating chemical reaction cycles under complex physiological conditions. Here, we develop organelle-mediated dissipative self-assembly of peptides in living cells fueled by GSH, via the design of a mitochondrion-targeting and redox-responsive hexapeptide. While the hexapeptide undergoes efficient redox-responsive self-assembly, the addition of GSH into the peptide solution in the presence of mitochondrion-biomimetic liposomes containing hydrogen peroxide allows for transient assembly of peptides. Internalization of the peptide by LPS-stimulated macrophages leads to the self-assembly of the peptide driven by GSH reduction and the association of the peptide assemblies with mitochondria. The association facilitates reversible oxidation of the reduced peptide by mitochondrion-residing ROS and thereby dissociates the peptide from mitochondria to re-enter the cytoplasm for GSH reduction. The metastable peptide-mitochondrion complexes prevent the thermodynamically equilibrated self-assembly, thus establishing dissipative assembly of peptides in stimulated macrophages. The entire dissipative self-assembling process allows for elimination of elevated ROS and decrease of pro-inflammatory cytokine expression. Creating dissipative self-assembling systems assisted by internal structures provides new avenues for the development of living materials or medical agents in the future.


Assuntos
Mitocôndrias , Peptídeos , Espécies Reativas de Oxigênio , Peptídeos/química
2.
J Am Chem Soc ; 146(15): 10753-10766, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38578841

RESUMO

Proteolysis targeting chimera (PROTAC) technology is an innovative strategy for cancer therapy, which, however, suffers from poor targeting delivery and limited capability for protein of interest (POI) degradation. Here, we report a strategy for the in situ formulation of antineoplastic Supra-PROTACs via intracellular sulfatase-responsive assembly of peptides. Coassembling a sulfated peptide with two ligands binding to ubiquitin VHL and Bcl-xL leads to the formation of a pro-Supra-PROTAC, in which the ratio of the two ligands is rationally optimized based on their protein binding affinity. The resulting pro-Supra-PROTAC precisely undergoes enzyme-responsive assembly into nanofibrous Supra-PROTACs in cancer cells overexpressing sulfatase. Mechanistic studies reveal that the pro-Supra-PROTACs selectively cause apparent cytotoxicity to cancer cells through the degradation of Bcl-xL and the activation of caspase-dependent apoptosis, during which the rationally optimized ligand ratio improves the bioactivity for POI degradation and cell death. In vivo studies show that in situ formulation enhanced the tumor accumulation and retention of the pro-Supra-PROTACs, as well as the capability for inhibiting tumor growth with excellent biosafety when coadministrating with chemodrugs. Our findings provide a new approach for enzyme-regulated assembly of peptides in living cells and the development of PROTACs with high targeting delivering and POI degradation efficiency.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Quimera de Direcionamento de Proteólise , Antineoplásicos/farmacologia , Sulfatases , Proteólise , Peptídeos , Ubiquitina-Proteína Ligases
3.
J Am Chem Soc ; 146(34): 24177-24187, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39140408

RESUMO

Despite significant progress achieved in artificial self-sorting in solution, operating self-sorting in the body remains a considerable challenge. Here, we report an in vivo self-sorting peptide system via an in situ assembly evolution for combined cancer therapy. The peptide E3C16-SS-EIY consists of two disulfide-connected segments, E3C16SH and SHEIY, capable of independent assembly into twisted or flat nanoribbons. While E3C16-SS-EIY assembles into nanorods, exposure to glutathione (GSH) leads to the conversion of the peptide into E3C16SH and SHEIY, thus promoting in situ evolution from the nanorods into self-sorted nanoribbons. Furthermore, incorporation of two ligand moieties targeting antiapoptotic protein XIAP and organellar endoplasmic reticulum (ER) into the self-sorted nanoribbons allows for simultaneous inhibition of XIAP and accumulation surrounding ER. This leads to the cytotoxicity toward the cancer cells with elevated GSH levels, through activating caspase-dependent apoptosis and inducing ER dysfunction. In vivo self-sorting of E3C16-SS-EIY decorated with ligand moieties is thoroughly validated by tissue studies. Tumor-bearing mouse experiments confirm the therapeutic efficacy of the self-sorted assemblies for inhibiting tumor growth, with excellent biosafety. Our findings demonstrate an efficient approach to develop in vivo self-sorting systems and thereby facilitating in situ formulation of biomedical agents.


Assuntos
Peptídeos , Humanos , Animais , Peptídeos/química , Peptídeos/farmacologia , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Glutationa/química , Glutationa/metabolismo , Linhagem Celular Tumoral , Nanotubos/química
4.
Biomacromolecules ; 25(4): 2497-2508, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38478850

RESUMO

Morphology-transformational self-assembly of peptides allows for manipulation of the performance of nanostructures and thereby advancing the development of biomaterials. Acceleration of the morphological transformation process under a biological microenvironment is important to efficiently implement the tailored functions in living systems. Herein, we report redox-regulated in situ seed-induced assembly of peptides via design of two co-assembled bola-amphiphiles serving as a redox-resistant seed and a redox-responsive assembly monomer, respectively. Both of the peptides are able to independently assemble into nanoribbons, while the seed monomer exhibits stronger assembling propensity. The redox-responsive monomer undergoes morphological transformation from well-defined nanoribbons to nanoparticles. Kinetics studies validate the role of the assembled inert monomer as the seeds in accelerating the assembly of the redox-responsive monomer. Alternative addition of oxidants and reductants into the co-assembled monomers promotes the redox-regulated assembly of the peptides facilitated by the in situ-formed seeds. The reduction-induced assembly of the peptide could also be accelerated by in situ-formed seeds in cancer cells with a high level of reductants. Our findings demonstrate that through precisely manipulating the assembling propensity of co-assembled monomers, the in situ seed-induced assembly of peptides could be achieved. Combining the rapid assembly kinetics of the seed-induced assembly with the common presence of redox agents in a biological microenvironment, this strategy potentially offers a new method for developing biomedical materials in living systems.


Assuntos
Nanoestruturas , Nanotubos de Carbono , Substâncias Redutoras , Peptídeos/química , Nanoestruturas/química , Materiais Biocompatíveis , Oxirredução
5.
Angew Chem Int Ed Engl ; 63(28): e202404703, 2024 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-38655625

RESUMO

Self-assembly in living cells represents one versatile strategy for drug delivery; however, it suffers from the limited precision and efficiency. Inspired by viral traits, we here report a cascade targeting-hydrolysis-transformation (THT) assembly of glycosylated peptides in living cells holistically resembling viral infection for efficient cargo delivery and combined tumor therapy. We design a glycosylated peptide via incorporating a ß-galactose-serine residue into bola-amphiphilic sequences. Co-assembling of the glycosylated peptide with two counterparts containing irinotecan (IRI) or ligand TSFAEYWNLLSP (PMI) results in formation of the glycosylated co-assemblies SgVEIP, which target cancer cells via ß-galactose-galectin-1 association and undergo galactosidase-induced morphological transformation. While GSH-reduction causes release of IRI from the co-assemblies, the PMI moieties release p53 and facilitate cell death via binding with protein MDM2. Cellular experiments show membrane targeting, endo-/lysosome-mediated internalization and in situ formation of nanofibers in cytoplasm by SgVEIP. This cascade THT process enables efficient delivery of IRI and PMI into cancer cells secreting Gal-1 and overexpressing ß-galactosidase. In vivo studies illustrate enhanced tumor accumulation and retention of the glycosylated co-assemblies, thereby suppressing tumor growth. Our findings demonstrate an in situ assembly strategy mimicking viral infection, thus providing a new route for drug delivery and cancer therapy in the future.


Assuntos
Sistemas de Liberação de Medicamentos , Glicopeptídeos , Humanos , Glicopeptídeos/química , Glicopeptídeos/metabolismo , Animais , Viroses/tratamento farmacológico , Viroses/metabolismo , Irinotecano/química , Irinotecano/farmacologia , Camundongos , Linhagem Celular Tumoral
6.
Chembiochem ; 24(3): e202200497, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36278304

RESUMO

In situ self-assembly of peptides into well-defined nanostructures represents one of versatile strategies for creation of bioactive materials within living cells with great potential in disease diagnosis and treatment. The intimate relationship between amino acid sequences and the assembling propensity of peptides has been thoroughly elucidated over the past few decades. This has inspired development of various controllable self-assembling peptide systems based on stimuli-responsive naturally occurring or non-canonical amino acids, including redox-, pH-, photo-, enzyme-responsive amino acids. This review attempts to summarize the recent progress achieved in manipulating in situ self-assembly of peptides by controllable reactions occurring to amino acids. We will highlight the systems containing non-canonical amino acids developed in our laboratory during the past few years, primarily including acid/enzyme-responsive 4-aminoproline, redox-responsive (seleno)methionine, and enzyme-responsive 2-nitroimidazolyl alanine. Utilization of the stimuli-responsive assembling systems in creation of bioactive materials will be specifically introduced to emphasize their advantages for addressing the concerns lying in disease theranostics. Eventually, we will provide the perspectives for the further development of stimulus-responsive amino acids and thereby demonstrating their great potential in development of next-generation biomaterials.


Assuntos
Aminoácidos , Nanoestruturas , Aminoácidos/química , Peptídeos/química , Sequência de Aminoácidos , Nanoestruturas/química , Materiais Biocompatíveis/química
7.
Macromol Rapid Commun ; 44(23): e2300308, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37462116

RESUMO

Bio-inspired design of peptides represents one facile strategy for development of supramolecular monomers for self-assembly into well-defined nanostructures. Inspired by methylation of arginine during post-translational modification for manipulating protein functions, herein, the controllable self-assembly of peptides via rational incorporation of methylated arginine residues into bola-amphiphilic peptides is reported. A series of bola-amphiphilic peptides are designed and synthesized either containing natural arginine or methylated arginine and investigate the influence of arginine methylation on peptide assembly. This study finds that incorporation of symmetrically di-methylated arginine into oppositely charged hexapeptide hex-SDMAE leads to distinct assembling performance compare to natural peptide hex-RE. The findings demonstrate that the methylation of rationally designed peptide sequences allows for regulation of self-assembly of peptides, thus implying the great potential of arginine methylation in establishing controllable peptide assembling systems and creating in situ formulation of biomedical materials in the future.


Assuntos
Arginina , Peptídeos , Arginina/química , Peptídeos/química , Proteínas , Processamento de Proteína Pós-Traducional , Metilação
8.
Angew Chem Int Ed Engl ; 62(49): e202314578, 2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-37870078

RESUMO

The presence of disordered region or large interacting surface within proteins significantly challenges the development of targeted drugs, commonly known as the "undruggable" issue. Here, we report a heterogeneous peptide-protein assembling strategy to selectively phosphorylate proteins, thereby activating the necroptotic signaling pathway and promoting cell necroptosis. Inspired by the structures of natural necrosomes formed by receptor interacting protein kinases (RIPK) 1 and 3, the kinase-biomimetic peptides are rationally designed by incorporating natural or D -amino acids, or connecting D -amino acids in a retro-inverso (DRI) manner, leading to one RIPK3-biomimetic peptide PR3 and three RIPK1-biomimetic peptides. Individual peptides undergo self-assembly into nanofibrils, whereas mixing RIPK1-biomimetic peptides with PR3 accelerates and enhances assembly of PR3. In particular, RIPK1-biomimetic peptide DRI-PR1 exhibits reliable binding affinity with protein RIPK3, resulting in specific cytotoxicity to colon cancer cells that overexpress RIPK3. Mechanistic studies reveal the increased phosphorylation of RIPK3 induced by RIPK1-biomimetic peptides, elucidating the activation of the necroptotic signaling pathway responsible for cell death without an obvious increase in secretion of inflammatory cytokines. Our findings highlight the potential of peptide-protein hybrid aggregation as a promising approach to address the "undruggable" issue and provide alternative strategies for overcoming cancer resistance in the future.


Assuntos
Apoptose , Peptídeos , Apoptose/fisiologia , Morte Celular , Fosforilação , Peptídeos/farmacologia , Aminoácidos
9.
Plant J ; 108(5): 1346-1364, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34582078

RESUMO

Tiller angle is an important determinant of plant architecture in rice (Oryza sativa L.). Auxins play a critical role in determining plant architecture; however, the underlying metabolic and signaling mechanisms are still largely unknown. In this study, we have identified a member of the bZIP family of TGA class transcription factors, OsbZIP49, that participates in the regulation of plant architecture and is specifically expressed in gravity-sensing tissues, including the shoot base, nodes and lamina joints. Transgenic rice plants overexpressing OsbZIP49 displayed a tiller-spreading phenotype with reduced plant height and internode lengths. In contrast, CRISPR/Cas9-mediated knockout of OsbZIP49 resulted in a compact architecture. Follow-up studies indicated that the effects of OsbZIP49 on tiller angles are mediated through changes in shoot gravitropic responses. Additionally, we provide evidence that OsbZIP49 activates the expression of indole-3-acetic acid-amido synthetases OsGH3-2 and OsGH3-13 by directly binding to TGACG motifs located within the promoters of both genes. Increased GH3-catalyzed conjugation of indole-3-acetic acid (IAA) in rice transformants overexpressing OsbZIP49 resulted in the increased accumulation of IAA-Asp and IAA-Glu, and a reduction in local free auxin, tryptamine and IAA-Glc levels. Exogenous IAA or naphthylacetic acid (NAA) partially restored shoot gravitropic responses in OsbZIP49-overexpressing plants. Knockout of OsbZIP49 led to reduced expression of both OsGH3-2 and OsGH3-13 within the shoot base, and increased accumulation of IAA and increased OsIAA20 expression levels were observed in transformants following gravistimulation. Taken together, the present results reveal the role transcription factor OsbZIP49 plays in determining plant architecture, primarily due to its influence on local auxin homeostasis.


Assuntos
Ácidos Indolacéticos/metabolismo , Oryza/genética , Reguladores de Crescimento de Plantas/metabolismo , Proteínas de Plantas/metabolismo , Expressão Gênica , Técnicas de Inativação de Genes , Gravitropismo , Homeostase , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Oryza/crescimento & desenvolvimento , Fenótipo , Proteínas de Plantas/genética , Plantas Geneticamente Modificadas , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
J Am Chem Soc ; 144(21): 9312-9323, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35587998

RESUMO

Self-sorting is a common phenomenon in eukaryotic cells and represents one of the versatile strategies for the formation of advanced functional materials; however, developing artificial self-sorting assemblies within living cells remains challenging. Here, we report on the GSH-responsive in situ self-sorting peptide assemblies within cancer cells for simultaneous organelle targeting to promote combinatorial organelle dysfunction and thereby cell death. The self-sorting system was created via the design of two peptides E3C16E6 and EVMSeO derived from lipid-inspired peptide interdigitating amphiphiles and peptide bola-amphiphiles, respectively. The distinct organization patterns of the two peptides facilitate their GSH-induced self-sorting into isolated nanofibrils as a result of cleavage of disulfide-connected hydrophilic domains or reduction of selenoxide groups. The GSH-responsive in situ self-sorting in the peptide assemblies within HeLa cells was directly characterized by super-resolution structured illumination microscopy. Incorporation of the thiol and ER-targeting groups into the self-sorted assemblies endows their simultaneous targeting of endoplasmic reticulum and Golgi apparatus, thus leading to combinatorial organelle dysfunction and cell death. Our results demonstrate the establishment of the in situ self-sorting peptide assemblies within living cells, thus providing a unique platform for drug targeting delivery and an alternative strategy for modulating biological processes in the future.


Assuntos
Complexo de Golgi , Peptídeos , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Peptídeos/química , Transporte Proteico
11.
J Am Chem Soc ; 144(15): 6907-6917, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35388694

RESUMO

Enzyme-regulated in situ self-assembly of peptides represents one versatile strategy in the creation of theranostic agents, which, however, is limited by the strong dependence on enzyme overexpression. Herein, we reported the self-amplifying assembly of peptides precisely in macrophages associated with enzyme expression for improving the anti-inflammatory efficacy of conventional drugs. The self-amplifying assembling system was created via coassembling an enzyme-responsive peptide with its derivative functionalized with a protein ligand. Reduction of the peptides by the enzyme NAD(P)H quinone dehydrogenase 1 (NQO1) led to the formation of nanofibers with high affinity to the protein, thereby facilitating NQO1 expression. The improved NQO1 level conversely promoted the assembly of the peptides into nanofibers, thus establishing an amplifying relationship between the peptide assembly and the NQO1 expression in macrophages. Utilization of the amplifying assembling system as vehicles for drug dexamethasone allowed for its passive targeting delivery to acute injured lungs. Both in vitro and in vivo studies confirmed the capability of the self-amplifying assembling system to enhance the anti-inflammatory efficacy of dexamethasone via simultaneous alleviation of the reactive oxygen species side effect and downregulation of proinflammatory cytokines. Our findings demonstrate the manipulation of the assembly of peptides in living cells with a regular enzyme level via a self-amplification process, thus providing a unique strategy for the creation of supramolecular theranostic agents in living cells.


Assuntos
Nanofibras , Peptídeos , Dexametasona , Ligantes , Macrófagos/metabolismo , Nanofibras/química , Peptídeos/química
12.
J Nanobiotechnology ; 20(1): 340, 2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35858873

RESUMO

Practice of tumor-targeted suicide gene therapy is hampered by unsafe and low efficient delivery of plasmid DNA (pDNA). Using HIV-Tat-derived peptide (Tat) to non-covalently form Tat/pDNA complexes advances the delivery performance. However, this innovative approach is still limited by intracellular delivery efficiency and cell-cycle status. In this study, Tat/pDNA complexes were further condensed into smaller, nontoxic nanoparticles by Ca2+ addition. Formulated Tat/pDNA-Ca2+ nanoparticles mainly use macropinocytosis for intercellular delivery, and their macropinocytic uptake was persisted in mitosis (M-) phase and highly activated in DNA synthesis (S-) phase of cell-cycle. Over-expression or phosphorylation of a mitochondrial chaperone, 75-kDa glucose-regulated protein (GRP75), promoted monopolar spindle kinase 1 (MPS1)-controlled centrosome duplication and cell-cycle progress, but also driven cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles. Further in vivo molecular imaging based on DF (Fluc-eGFP)-TF (RFP-Rluc-HSV-ttk) system showed that Tat/pDNA-Ca2+ nanoparticles exhibited highly suicide gene therapy efficiency in mouse model xenografted with human ovarian cancer. Furthermore, arresting cell-cycle at S-phase markedly enhanced delivery performance of Tat/pDNA-Ca2+ nanoparticles, whereas targeting GRP75 reduced their macropinocytic delivery. More importantly, in vivo targeting GRP75 combined with cell-cycle or macropinocytosis inhibitors exhibited distinct suicide gene therapy efficiency. In summary, our data highlight that mitochondrial chaperone GRP75 moonlights as a biphasic driver underlying cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles in ovarian cancer.


Assuntos
Nanopartículas , Neoplasias Ovarianas , Animais , Cálcio , DNA/química , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Proteínas de Choque Térmico HSP70 , Humanos , Proteínas de Membrana , Camundongos , Nanopartículas/química , Neoplasias Ovarianas/terapia , Plasmídeos , Transfecção
13.
Nano Lett ; 21(13): 5730-5737, 2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34142834

RESUMO

Mitochondrion-targeting therapy exhibits great potential in cancer therapy but significantly suffers from limited therapeutic efficiency. Here we report on mitochondrion-targeting supramolecular antagonist-inducing tumor cell death via simultaneously promoting cellular apoptosis and preventing survival. The supramolecular antagonist was created via coassembly of a mitochondrion-targeting pentapeptide with its two derivatives functionalized with a BH3 domain or the drug camptothecin (CPT). While drug CPT released from the antagonist induced cellular apoptosis via decreasing the mitochondrial membrane potential, the BH3 domain prevented cellular survival through facilitating the association between the supramolecular antagonists and antiapoptotic proteins, thereby initiating mitochondrial permeabilization. Both in vitro and in vivo studies confirmed the combinatorial therapeutic effect arising from the BH3 domain and CPT drug within the supramolecular antagonist on cell death and thereby inhibiting tumor growth. Our findings demonstrate an efficient combinatorial mechanism for mitochondrial dysfunction, thus potentially serving as novel organelle-targeting medicines.


Assuntos
Apoptose , Camptotecina , Camptotecina/farmacologia , Mitocôndrias
14.
J Am Chem Soc ; 143(34): 13854-13864, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34410694

RESUMO

Design of endogenous stimuli-responsive amino acids allows for precisely modulating proteins or peptides under a biological microenvironment and thereby regulating their performance. Herein we report a noncanonical amino acid 2-nitroimidazol-1-yl alanine and explore its functions in creation of the nitroreductase (NTR)-responsive peptide-based supramolecular probes for efficient hypoxia imaging. On the basis of the reduction potential of the nitroimidazole unit, the amino acid was synthesized via the Mitsunobu reaction between 2-nitroimidazole and a serine derivate. We elucidated the relationship between the NTR-responsiveness of the amino acid and the structural feature of peptides involving a series of peptides. This eventually facilitates development of aromatic peptides undergoing NTR-responsive self-assembly by rationally optimizing the sequences. Due to the intrinsic role of 2-nitroimidazole in the fluorescence quench, we created a morphology-transformable supramolecular probe for imaging hypoxic tumor cells based on NTR reduction. We found that the resulting supramolecular probes penetrated into solid tumors, thus allowing for efficient fluorescence imaging of tumor cells in hypoxic regions. Our findings demonstrate development of a readily synthesized and versatile amino acid with exemplified properties in creating fluorescent peptide nanostructures responsive to a biological microenvironment, thus providing a powerful toolkit for synthetic biology and development of novel biomaterials.


Assuntos
Aminoácidos/metabolismo , Peptídeos/metabolismo , Alanina/química , Alanina/metabolismo , Aminoácidos/química , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Corantes Fluorescentes/química , Camundongos , Microscopia Confocal , Nitroimidazóis/química , Nitrorredutases/metabolismo , Imagem Óptica , Peptídeos/química , Transplante Homólogo
15.
Nano Lett ; 19(11): 7965-7976, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31596096

RESUMO

Nanomedicines have been demonstrated as promising strategies for cancer therapy due to the advantages in pharmacokinetics and drug targeting delivery to tumor tissues. However, creation of delivery platforms able to intrinsically and spatially optimize drug cellular uptake during the entire delivering process remain challenging. To address this challenge, here we report on tumor microenvironment-adaptable self-assembly (TMAS) of pentapeptides regulated by the pH-sensitive cis/trans isomerization of 4-amino-proline (Amp) amide bonds for enhanced drug delivery and photodynamic therapeutic (PDT) efficacy. We found that decreasing solution pH led to the cis → trans isomerization of Amp amide bonds, thus promoting reversible self-assembly of pentapeptide FF-Amp-FF (AmpF) into superhelices and nanoparticles upon alternating exposure to neutral and mild acidic conditions. Co-assembly of peptide AmpF with its derivative containing a photosensitizer Chlorin e6 (AmpF-C) allows for creation of TMAS systems undergoing a morphological transition adaptable to the pH gradient present in cellular uptake pathway. Ex vivo studies revealed that TMAS nanomedicines prolonged circulation in the animal body and improved accumulation at tumor sites compared to morphology-persistent nanomedicines. In addition to the optimized cellular uptake, the morphological transition of TMAS into nanofibers in cytoplasm caused an enhanced intracellular ROS level compared to nanoparticle counterparts, thus leading to a lowered half lethal dose value for cancer cells. The combined advantages of TMAS eventually allowed in vivo PDT therapy for significant inhibition of tumor growth, thus demonstrating the improved drug delivery efficiency and therapeutic efficacy of TMAS systems toward new-generation nanomedicines.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Preparações de Ação Retardada/química , Oligopeptídeos/química , Fármacos Fotossensibilizantes/administração & dosagem , Porfirinas/administração & dosagem , Prolina/química , Animais , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Clorofilídeos , Feminino , Isomerismo , Camundongos , Nanoestruturas/química , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos
16.
Langmuir ; 35(13): 4710-4717, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30836752

RESUMO

Precise incorporation of functional residues into sequences allows for tailoring the noncovalent interactions between peptides to guide their self-assembly into well-defined nanostructures, thus facilitating creation of artificial functional materials resembling natural systems. Here, we report on the self-assembly of dipeptides consisting of one fluorinated phenylalanine unit (Z residue) and one natural aromatic residue into laminated nanofibers predominately driven by polar-π interactions. On the basis of characterizations using transmission electron microscopy, scanning electron microscopy, atomic force microscopy, circular dichroism, Fourier transform infrared spectroscopy, and thioflavin T binding assay, we found that the face-centered stacking pattern of the dipeptides FZ, ZF, and ZY stabilized by the polar-π interactions and antiparallel ß-sheet H-bonding interactions led to lamination of nanofibers and formation of ribbonlike nanostructures. Our findings demonstrate that incorporation of fluorinated aromatic units into short peptides not only promotes of polar-π interactions as alternative self-assembling driving forces but also governs the organizing pattern of peptides, thus benefiting creation of well-defined peptide nanostructures.

17.
J Am Chem Soc ; 139(23): 7823-7830, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28571316

RESUMO

Co-assembly of binary systems driven by specific non-covalent interactions can greatly expand the structural and functional space of supramolecular nanostructures. We report here on the self-assembly of peptide amphiphiles and fatty acids driven primarily by anion-π interactions. The peptide sequences investigated were functionalized with a perfluorinated phenylalanine residue to promote anion-π interactions with carboxylate headgroups in fatty acids. These interactions were verified here by NMR and circular dichroism experiments as well as investigated using atomistic simulations. Positioning the aromatic units close to the N-terminus of the peptide backbone near the hydrophobic core of cylindrical nanofibers leads to strong anion-π interactions between both components. With a low content of dodecanoic acid in this position, the cylindrical morphology is preserved. However, as the aromatic units are moved along the peptide backbone away from the hydrophobic core, the interactions with dodecanoic acid transform the cylindrical supramolecular morphology into ribbon-like structures. Increasing the ratio of dodecanoic acid to PA leads to either the formation of large vesicles in the binary systems where the anion-π interactions are strong, or a heterogeneous mixture of assemblies when the peptide amphiphiles associate weakly with dodecanoic acid. Our findings reveal how co-assembly involving designed specific interactions can drastically change supramolecular morphology and even cross from nano to micro scales.


Assuntos
Ácidos Láuricos/química , Lipídeos/química , Nanoestruturas/química , Peptídeos/química , Tensoativos/química , Ânions/química , Interações Hidrofóbicas e Hidrofílicas , Substâncias Macromoleculares/química , Conformação Molecular , Simulação de Dinâmica Molecular
18.
Nano Lett ; 16(11): 6967-6974, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27797531

RESUMO

Asymmetry in chemical structure or shape at molecular, nanoscale, or microscopic levels is essential to a vast number of functionalities in both natural and artificial systems. Bottom-up approaches to create asymmetric supramolecular nanostructures are considered promising but this strategy suffers from the potentially dynamic nature of noncovalent interactions. We report here on supramolecular self-assembly of asymmetric peptide amphiphiles consisting of two different molecularly linked domains. We found that strong noncovalent interactions and a high degree of internal order among the asymmetric amphiphiles lead to nanoribbons with asymmetric faces due to the preferential self-association of the two domains. The capture of gold nanoparticles on only one face of the nanoribbons demonstrates symmetry breaking in these supramolecular structures.


Assuntos
Nanotubos de Carbono/química , Peptídeos/química , Cloreto de Cálcio/química , Elétrons , Ouro/química , Interações Hidrofóbicas e Hidrofílicas , Nanopartículas Metálicas/química , Tamanho da Partícula , Conformação Proteica , Multimerização Proteica , Reologia , Propriedades de Superfície , Termodinâmica
19.
J Am Chem Soc ; 138(39): 12997-13005, 2016 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-27598007

RESUMO

Investigating and deciphering the dynamics of photoswitchable foldamers provides a detailed understanding of their photoinduced conformational transitions, resembling similar processes in photoresponsive biomacromolecules. We studied the ultrafast dynamics of the photoisomerization of azobenzene moieties embedded in a foldamer backbone and the resulting conformational helix-coil transition by time-resolved femtosecond/picosecond pump-probe spectroscopy in the visible and infrared region. During E → Z photoisomerization of the azobenzenes, the complexity of the photoinduced conformational transition of the pentameric foldamer 105 is reflected in distinct spectral characteristics and a 2-fold slower decay of the excited-state absorption bands compared to the monomer M (τ4,foldamer = 20 ps, τ4,monomer = 9 ps). Time-resolved IR experiments reveal the vibrational features of the monomer and the foldamer after photoexcitation, with an additional time constant for the foldamer (τ = 150 ps), indicating the initial steps of unfolding of the helical conformation, which are supported by density functional theory calculations. Our results record the overall sequence of photoinduced structural changes in the foldamer, starting from the initial ultrafast isomerization of the azobenzene unit(s) and ending with the complete unfolding on a later time scale. From our experiments, we could gain insight into the coupling of primary photoisomerization events ("cause") and secondary unfolding processes ("effect") in these oligoazobenzene foldamers.

20.
ACS Appl Mater Interfaces ; 16(35): 45821-45829, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39177358

RESUMO

In situ self-assembly in living systems is referred to as the processes that regulate assembly by stimuli-responsive reactions at target sites under physiological conditions. Due to the advantages of precisely forming well-defined nanostructures at pathological lesions, in situ-formed assemblies with tailored bioactivity are promising for the development of next-generation biomedical agents. In this Perspective, we summarize the progress of in situ self-assembly of peptides in living cells with an emphasis on the state-of-the-art strategies regulating assembly processes, establishing complexity within assembly systems, and exploiting their applications in biomedicines. We also provide our forward conceiving perspectives on the challenges in the development of in situ assembly in living cells to demonstrate its great potential in creating biomaterials for healthcare in the future.


Assuntos
Materiais Biocompatíveis , Humanos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/farmacologia , Nanoestruturas/química , Peptídeos/química , Peptídeos/síntese química , Animais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa