Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(33): e2405454121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39106310

RESUMO

Regeneration of hyaline cartilage in human-sized joints remains a clinical challenge, and it is a critical unmet need that would contribute to longer healthspans. Injectable scaffolds for cartilage repair that integrate both bioactivity and sufficiently robust physical properties to withstand joint stresses offer a promising strategy. We report here on a hybrid biomaterial that combines a bioactive peptide amphiphile supramolecular polymer that specifically binds the chondrogenic cytokine transforming growth factor ß-1 (TGFß-1) and crosslinked hyaluronic acid microgels that drive formation of filament bundles, a hierarchical motif common in natural musculoskeletal tissues. The scaffold is an injectable slurry that generates a porous rubbery material when exposed to calcium ions once placed in cartilage defects. The hybrid material was found to support in vitro chondrogenic differentiation of encapsulated stem cells in response to sustained delivery of TGFß-1. Using a sheep model, we implanted the scaffold in shallow osteochondral defects and found it can remain localized in mechanically active joints. Evaluation of resected joints showed significantly improved repair of hyaline cartilage in osteochondral defects injected with the scaffold relative to defects injected with the growth factor alone, including implantation in the load-bearing femoral condyle. These results demonstrate the potential of the hybrid biomimetic scaffold as a niche to favor cartilage repair in mechanically active joints using a clinically relevant large-animal model.


Assuntos
Condrogênese , Alicerces Teciduais , Fator de Crescimento Transformador beta1 , Animais , Alicerces Teciduais/química , Ovinos , Fator de Crescimento Transformador beta1/metabolismo , Condrogênese/efeitos dos fármacos , Polímeros/química , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Cartilagem Articular/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Engenharia Tecidual/métodos , Humanos , Materiais Biocompatíveis/química , Condrócitos/efeitos dos fármacos , Cartilagem Hialina/metabolismo
2.
ACS Appl Bio Mater ; 7(8): 5553-5565, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39042039

RESUMO

Chirality plays a crucial role in the self-assembly of biomolecules in nature. Peptides show chirality-dependent conformation and self-assembly. Lipidation of peptides occurs in vivo and has recently been exploited in designed conjugates to drive self-assembly and enhance bioactivity. Here, a library of pH-responsive homochiral and heterochiral lipidated tripeptides has been designed. The designed lipopeptides comprise homochiral C16-YKK or C16-WKK (where all the amino acids are l-isomers), and two heterochiral conjugates C16-Ykk and C16-Wkk (where the two lysines are d-isomers). The self-assembly of all the synthesized lipopeptides in aqueous solution was examined using a combination of spectroscopic methods along with cryogenic-transmission electron microscopy (cryo-TEM) and small-angle X-ray scattering (SAXS). Interestingly, it was observed that at acidic pH all the lipopeptides self-assemble into micelles, whereas at basic pH the homochiral lipopeptides self-assemble into nanofibers, whereas the heterochiral lipopeptides self-assemble into nanotapes and nanotubes. A pH switch was demonstrated using a thioflavin T fluorescence probe of ß-sheet structure present in the extended structures at pH 8. We demonstrate that both chirality and pH in lipopeptides influence the self-assembly behavior of the model tripeptides, which also show promising bioactivity. Good cytocompatibility is observed in hemolytic assays and antimicrobial activity against both Gram-negative and Gram-positive bacteria is shown through the determination of minimum inhibition concentration (MIC) and minimum bactericidal concentration (MBC) values and live/dead bacteria staining assay.


Assuntos
Antibacterianos , Materiais Biocompatíveis , Lipopeptídeos , Teste de Materiais , Testes de Sensibilidade Microbiana , Nanoestruturas , Tamanho da Partícula , Concentração de Íons de Hidrogênio , Lipopeptídeos/química , Lipopeptídeos/farmacologia , Nanoestruturas/química , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/síntese química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/síntese química , Estrutura Molecular , Estereoisomerismo , Humanos
3.
Mol Pharm ; 21(9): 4498-4509, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39069731

RESUMO

Recent emphasis on the design of drug delivery systems typically involves the effective transport of a pharmaceutical substance to the disease site with the desired therapeutic efficacy and minimal cytotoxicity. Organelle-targeted peptides have become an integral part of designing an important class of prodrug/prodrug assemblies for new supramolecular therapeutics owing to their favorable biocompatibility, synthetic ease, tunability of their aggregation behavior, and desired functionalization for site-specificity. However, it is still limited due to the low selectivity. We designed a folic acid-functionalized ß-cyclodextrin (FA-CD) as a delivery platform for specific and selective delivery of organelle-targeted (such as microtubule, lysosome, and mitochondria) peptide chemotherapeutics to the folate receptor (FR) overexpressing cancer cell lines. Low toxicity was found for the FA-CD and organelle-targeted peptide inclusion complex in FR-negative normal cells, but superior inhibition of tumor growth with no in vivo toxicity was found for the inclusion complex in the xenograft tumor model.


Assuntos
Sistemas de Liberação de Medicamentos , Ácido Fólico , Peptídeos , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Ciclodextrinas , Ácido Fólico/química , Humanos , beta-Ciclodextrinas/química , Animais , Camundongos , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/química , Peptídeos/farmacologia , Linhagem Celular Tumoral , Organelas/efeitos dos fármacos , Organelas/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/administração & dosagem , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/metabolismo , Receptores de Folato com Âncoras de GPI/metabolismo , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pró-Fármacos/administração & dosagem , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Feminino
4.
Proc Natl Acad Sci U S A ; 121(16): e2321498121, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38593077

RESUMO

In recent decades, peptide amphiphiles (PAs) have established themselves as promising self-assembling bioinspired materials in a wide range of medical fields. Herein, we report a dual-therapeutic system constituted by an antimicrobial PA and a cylindrical protease inhibitor (LJC) to achieve broad antimicrobial spectrum and to enhance therapeutic efficacy. We studied two strategies: PA-LJC nanostructures (Encapsulation) and PA nanostructures + free LJC (Combination). Computational modeling using a molecular theory for amphiphile self-assembly captures and explains the morphology of PA-LJC nanostructures and the location of encapsulated LJC in agreement with transmission electron microscopy and two-dimensional (2D) NMR observations. The morphology and release profile of PA-LJC assemblies are strongly correlated to the PA:LJC ratio: high LJC loading induces an initial burst release. We then evaluated the antimicrobial activity of our nanosystems toward gram-positive and gram-negative bacteria. We found that the Combination broadens the spectrum of LJC, reduces the therapeutic concentrations of both agents, and is not impacted by the inoculum effect. Further, the Encapsulation provides additional benefits including bypassing water solubility limitations of LJC and modulating the release of this molecule. The different properties of PA-LJC nanostructures results in different killing profiles, and reduced cytotoxicity and hemolytic activity. Meanwhile, details in membrane alterations caused by each strategy were revealed by various microscopy and fluorescent techniques. Last, in vivo studies in larvae treated by the Encapsulation strategy showed better antimicrobial efficacy than polymyxin B. Collectively, this study established a multifunctional platform using a versatile PA to act as an antibiotic, membrane-penetrating assistant, and slow-release delivery vehicle.


Assuntos
Anti-Infecciosos , Nanoestruturas , Antibacterianos/farmacologia , Antibacterianos/química , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Nanoestruturas/química
5.
Acta Biomater ; 177: 50-61, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-38331132

RESUMO

Cell therapies offer great promise in the treatment of diseases and tissue regeneration, but their clinical use has many challenges including survival, optimal performance in their intended function, or localization at sites where they are needed for effective outcomes. We report here on a method to coat a biodegradable matrix of biomimetic nanofibers on single cells that could have specific functions ranging from cell signaling to targeting and helping cells survive when used for therapies. The fibers are composed of peptide amphiphile (PA) molecules that self-assemble into supramolecular nanoscale filaments. The PA nanofibers were able to create a mesh-like coating for a wide range of cell lineages with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The targeting abilities of this system were assessed in vitro using human primary regulatory T (hTreg) cells coated with PAs displaying a vascular cell adhesion protein 1 (VCAM-1) targeting motif. This approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies. STATEMENT OF SIGNIFICANCE: Cell therapies hold great promise in the treatment of diseases and tissue regeneration, but their clinical use has been limited by cell survival, targeting, and function. We report here a method to coat single cells with a biodegradable matrix of biomimetic nanofibers composed of peptide amphiphile (PA) molecules. The nanofibers were able to coat cells, such as human primary regulatory T cells, with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies.


Assuntos
Nanofibras , Humanos , Nanofibras/química , Biomimética , Matriz Extracelular , Peptídeos/farmacologia , Peptídeos/química
6.
Curr Med Chem ; 31(11): 1332-1347, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37031390

RESUMO

During the last few decades, several efforts have been made towards developing biocompatible materials. Among them, peptide amphiphiles (PAs) constitute a novel nanotechnological strategy used in the field of biomedicine since they can provide tissue- specific binding and localization. PAs possess several regions combining hydrophobic and hydrophilic areas that are able to self-assemble in aqueous media, forming different tertiary nanostructures able to interact with cellular membranes. Moreover, these molecules can be tuned by incorporating collagen, lipids, or fluorescent markers. In addition, they can also be used as carriers in order to encapsulate active compounds for drug delivery showing promising features in this area. In this review, the self-assembled structures of PAs as well as their pharmacological applications have been summarized. Furthermore, their use as drug delivery systems has been highlighted and the latest advances in this field have been reviewed.


Assuntos
Nanoestruturas , Peptídeos , Humanos , Preparações Farmacêuticas , Peptídeos/farmacologia , Peptídeos/química , Nanoestruturas/química , Sistemas de Liberação de Medicamentos , Materiais Biocompatíveis
7.
Adv Healthc Mater ; 13(4): e2301364, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37947246

RESUMO

Retroviral gene delivery is the key technique for in vitro and ex vivo gene therapy. However, inefficient virion-cell attachment resulting in low gene transduction efficacy remains a major challenge in clinical applications. Adjuvants for ex vivo therapy settings need to increase transduction efficiency while being easily removed or degraded post-transduction to prevent the risk of venous embolism after infusing the transduced cells back to the bloodstream of patients, yet no such peptide system have been reported thus far. In this study, peptide amphiphiles (PAs) with a hydrophobic fatty acid and a hydrophilic peptide moiety that reveal enhanced viral transduction efficiency are introduced. The PAs form ß-sheet-rich fibrils that assemble into positively charged aggregates, promoting virus adhesion to the cell membrane. The block-type amphiphilic sequence arrangement in the PAs ensures efficient cell-virus interaction and biodegradability. Good biodegradability is observed for fibrils forming small aggregates and it is shown that via molecular dynamics simulations, the fibril-fibril interactions of PAs are governed by fibril surface hydrophobicity. These findings establish PAs as additives in retroviral gene transfer, rivalling commercially available transduction enhancers in efficiency and degradability with promising translational options in clinical gene therapy applications.


Assuntos
Técnicas de Transferência de Genes , Peptídeos , Humanos , Peptídeos/química , Terapia Genética , Adjuvantes Imunológicos
8.
Chemistry ; 30(7): e202303194, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37967312

RESUMO

Developing peptide-based materials with controlled morphology is a critical theme of soft matter research. Herein, we report the formation of a novel, patterned cross-ß structure formed by self-assembled C3 -symmetric peptide amphiphiles based on diphenylalanine and benzene-1,3,5-tricarboxamide (BTA). The cross-ß motif is an abundant structural element in amyloid fibrils and aggregates of fibril-forming peptides, including diphenylalanine. The incorporation of topological constraints on one edge of the diphenylalanine fragment limits the number of ß-strands in ß-sheets and leads to the creation of an unconventional offset-patterned cross-ß structure consisting of short 3×2 parallel ß-sheets stabilized by phenylalanine zippers. In the reported assembly, two patterned cross-ß structures bind parallel arrays of BTA stacks in a superstructure within a single-molecule-thick nanoribbon. In addition to a threefold network of hydrogen bonds in the BTA stack, each molecule becomes simultaneously bound by hydrogen bonds from three ß-sheets and four phenylalanine zippers. The diffuse layer of alkyl chains with terminal polar groups prevents the nanoribbons from merging and stabilizes cross-ß-structure in water. Our results provide a simple approach to the incorporation of novel patterned cross-ß motifs into supramolecular superstructures and shed light on the general mechanism of ß-sheet formation in C3 -symmetric peptide amphiphiles.


Assuntos
Amiloide , Peptídeos , Estrutura Secundária de Proteína , Peptídeos/química , Amiloide/química , Conformação Proteica em Folha beta , Fenilalanina
9.
Adv Colloid Interface Sci ; 322: 103033, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37931380

RESUMO

Self-assemblies of de novo designed short peptides at interface and in bulk solution provide potential platforms for developing applications in many medical and technological areas. However, characterising how bioinspired supramolecular nanostructures evolve with dynamic self-assembling processes and respond to different stimuli remains challenging. Neutron scattering technologies including small angle neutron scattering (SANS) and neutron reflection (NR) can be advantageous and complementary to other state-of-the-art techniques in tracing structural changes under different conditions. With more neutron sources now available, SANS and NR are becoming increasingly popular in studying self-assembling processes of diverse peptide and protein systems, but the difficulty in experimental manipulation and data analysis can deter beginners. This review will introduce the basic theory, general experimental setup and data analysis of SANS and NR, followed by provision of their applications in characterising interfacial and solution self-assemblies of representative peptides and proteins. SANS and NR are remarkably effective in determining the morphological features self-assembled short peptides, especially size and shape transitions as a result of either sequence changes or in response to environmental stimuli, demonstrating the unique capability of NR and SANS in unravelling the interactive processes. These examples highlight the potential of NR and SANS in supporting the development of novel short peptides and proteins as biopharmaceutical candidates in the fight against many diseases and infections that share common features of membrane interactive processes.


Assuntos
Peptídeos , Proteínas , Peptídeos/química , Proteínas/química , Nêutrons , Espalhamento a Baixo Ângulo
10.
Acta Biomater ; 171: 223-238, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37793600

RESUMO

Organoids are an emerging technology with great potential in human disease modelling, drug development, diagnosis, tissue engineering, and regenerative medicine. Organoids as 3D-tissue culture systems have gained special attention in the past decades due to their ability to faithfully recapitulate the complexity of organ-specific tissues. Despite considerable successes in culturing physiologically relevant organoids, their real-life applications are currently limited by challenges such as scarcity of an appropriate biomimetic matrix. Peptide amphiphiles (PAs) due to their well-defined chemistry, tunable bioactivity, and extracellular matrix (ECM)-like nanofibrous architecture represent an attractive material scaffold for organoids development. Using cerebral organoids (COs) as exemplar, we demonstrate the possibility to create bio-instructive hydrogels with tunable stiffness ranging from 0.69 kPa to 2.24 kPa to culture and induce COs growth. We used orthogonal chemistry involving oxidative coupling and supramolecular interactions to create two-component hydrogels integrating the bio-instructive activity and ECM-like nanofibrous architecture of a laminin-mimetic PAs (IKVAV-PA) and tunable crosslinking density of hyaluronic acid functionalized with tyramine (HA-Try). Multi-omics technology including transcriptomics, proteomics, and metabolomics reveals the induction and growth of COs in soft HA-Tyr hydrogels containing PA-IKVAV such that the COs display morphology and biomolecular signatures similar to those grown in Matrigel scaffolds. Our materials hold great promise as a safe synthetic ECM for COs induction and growth. Our approach represents a well-defined alternative to animal-derived matrices for the culture of COs and might expand the applicability of organoids in basic and clinical research. STATEMENT OF SIGNIFICANCE: Synthetic bio-instructive materials which display tissue-specific functionality and nanoscale architecture of the native extracellular matrix are attractive matrices for organoids development. These synthetic matrices are chemically defined and animal-free compared to current gold standard matrices such as Matrigel. Here, we developed hydrogel matrices with tunable stiffness, which incorporate laminin-mimetic peptide amphiphiles to grow and expand cerebral organoids. Using multi-omics tools, the present study provides exciting data on the effects of neuro-inductive cues on the biomolecular profiles of brain organoids.


Assuntos
Hidrogéis , Laminina , Animais , Humanos , Hidrogéis/farmacologia , Hidrogéis/química , Multiômica , Organoides , Peptídeos/farmacologia
11.
Nanomedicine (Lond) ; 18(17): 1095-1108, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37610088

RESUMO

Background: Corneal neovascularization is a sight-threatening disease. It can be treated using antiangiogenic and anti-inflammatory compounds. Therefore, atorvastatin (ATV) constitutes a suitable candidate to be administered topically. To attain suitable efficacy, ATV can be encapsulated into custom-developed nanocarriers such as peptide amphiphiles. Methods: Three peptide amphiphiles bearing one, two or four C16-alkyl groups (mC16-Tat47-57, dC16-Tat47-57 and qC16-Tat47-57) were synthesized, characterized and loaded with ATV. Drug release and ocular tolerance were assessed as well as anti-inflammatory and antiangiogenic properties. Results: ATV-qC16-Tat47-57 showed higher encapsulation efficiency than mC16-Tat47-57 and dC16-Tat47-57 and more defined nanostructures. ATV-qC16-Tat47-57 showed ATV prolonged release with suitable ocular tolerance. Moreover, ATV-qC16-Tat47-57 was antiangiogenic and prevented ocular inflammation. Conclusion: ATV-qC16-Tat47-57 constitutes a promising topical medication against corneal neovascularization.


Corneal neovascularization is an eye disease that affects over 1 million people every year and can lead to blindness. It is caused by inflammation and the unwanted formation of blood vessels in the eye. Current treatments for this disease are not fully effective. Atorvastatin (ATV) is one drug that has been partially successful at treating corneal neovascularization, but it does not stay in the eye long enough and does not mix well with the water-based environment of the eye. To overcome this, ATV was combined with three specially designed nanocarriers. These nanocarriers were peptides, short stretches of protein. They were designed to be amphiphilic, meaning that one section is hydrophilic (literally meaning 'water loving') and one section is hydrophobic ('water hating'). These peptide nanocarriers allowed ATV to stay in the water-based environment of the eye longer. The peptide with the most hydrophobic chains (qC16-Tat47-57) was able to carry more ATV than the other peptides and produced particles of a desired shape. ATV-qC16-Tat47-57 nanocarriers were found to release slowly. These nanocarriers were also found to prevent the development of new blood vessels on a membrane in a hen's egg used to mimic the eye. There was also no sign of irritation on this membrane or in the eyes of New Zealand rabbits. These results show ATV-qC16-Tat47-57 has a prolonged therapeutic effect, prevents the formation of new blood vessels and is tolerated in the eye. ATV-qC16-Tat47-57 is therefore potentially a more effective alternative to ATV treatment alone.


Assuntos
Neovascularização da Córnea , Humanos , Neovascularização da Córnea/tratamento farmacológico , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Olho , Liberação Controlada de Fármacos , Peptídeos
12.
Micromachines (Basel) ; 14(4)2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37421116

RESUMO

Synthetic peptides are promising structural and functional components of bioactive and tissue-engineering scaffolds. Here, we demonstrate the design of self-assembling nanofiber scaffolds based on peptide amphiphile (PA) molecules containing multi-functional histidine residues with trace metal (TM) coordination ability. The self-assembly of PAs and characteristics of PA nanofiber scaffolds along with their interaction with Zn, Cu, and Mn essential microelements were studied. The effects of TM-activated PA scaffolds on mammalian cell behavior, reactive oxygen species (ROS), and glutathione levels were shown. The study reveals the ability of these scaffolds to modulate adhesion, proliferation, and morphological differentiation of neuronal PC-12 cells, suggesting a particular role of Mn(II) in cell-matrix interaction and neuritogenesis. The results provide a proof-of-concept for the development of histidine-functionalized peptide nanofiber scaffolds activated with ROS- and cell-modulating TMs to induce regenerative responses.

13.
Chemistry ; 29(54): e202301678, 2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37358020

RESUMO

Biomaterials with improved biological features can be obtained by conjugating glycans to nanostructured peptides. Creating peptide-glycan chimeras requires superb chemoselectivity. We expedite access to such chimeras by merging peptide and glycan solid-phase syntheses employing a bifunctional monosaccharide. The concept was explored in the context of the on-resin generation of a model α(1→6)tetramannoside linked to peptides, lipids, steroids, and adamantane. Chimeras containing a ß(1→6)tetraglucoside and self-assembling peptides such as FF, FFKLVFF, and the amphiphile palmitoyl-VVVAAAKKK were prepared in a fully automated manner. The robust synthetic protocol requires a single purification step to obtain overall yields of about 20 %. The ß(1→6)tetraglucoside FFKLVFF chimera produces micelles rather than nanofibers formed by the peptide alone as judged by microscopy and circular dichroism. The peptide amphiphile-glycan chimera forms a disperse fiber network, creating opportunities for new glycan-based nanomaterials.

14.
ACS Appl Mater Interfaces ; 15(17): 20712-20725, 2023 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-37071430

RESUMO

Bioprinting is a precise layer-by-layer manufacturing technology utilizing biomaterials, cells, and sometimes growth factors for the fabrication of customized three-dimensional (3D) biological constructs. In recent years, it has gained considerable interest in various biomedical studies. However, the translational application of bioprinting is currently impeded by the lack in efficient techniques for blood vessel fabrications. In this report, by systematically studying the previously reported phenomenon, interfacial polyelectrolyte complexation, an efficient blood vessel bioprinting technique based on the phenomenon, was proposed and subsequently investigated. In this technique, anionic hyaluronate and cationic lysine-based peptide amphiphiles were placed concentrically to bioprint with human umbilical endothelial cells for the fabrication of biological tubular constructs. These constructs demonstrated clear vascular features, which made them highly resemble blood vessels. In addition, to optimize the bioactivity of the printed constructs, this report also, for the first time, studied peptide sequencing's effect on the biocompatibility of the polyelectrolyte-peptide amphiphile complex. All these studies conducted in the report are highly relevant and interesting for research in vascular structure fabrication, which will eventually be beneficial for translational application development of bioprinting.


Assuntos
Bioimpressão , Alicerces Teciduais , Humanos , Alicerces Teciduais/química , Polieletrólitos , Células Endoteliais , Bioimpressão/métodos , Impressão Tridimensional , Engenharia Tecidual/métodos
15.
Small ; 19(25): e2301063, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36932893

RESUMO

As an important noncovalent interaction, cation-π interaction plays an essential role in a broad area of biology and chemistry. Despite extensive studies in protein stability and molecular recognition, the utilization of cation-π interaction as a major driving force to construct supramolecular hydrogel remains uncharted. Here, a series of peptide amphiphiles are designed with cation-π interaction pairs that can self-assemble into supramolecular hydrogel under physiological condition. The influence of cation-π interaction is thoroughly investigated on peptide folding propensity, morphology, and rigidity of the resultant hydrogel. Computational and experimental results confirm that cation-π interaction could serve as a major driving force to trigger peptide folding, resultant ß-hairpin peptide self-assembled into fibril-rich hydrogel. Furthermore, the designed peptides exhibit high efficacy on cytosolic protein delivery. As the first case of using cation-π interactions to trigger peptide self-assembly and hydrogelation, this work provides a novel strategy to generate supramolecular biomaterials.


Assuntos
Hidrogéis , Peptídeos , Humanos , Hidrogéis/química , Peptídeos/química
16.
Adv Colloid Interface Sci ; 314: 102866, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36898186

RESUMO

Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.


Assuntos
Nanofibras , Nanoestruturas , Engenharia Tecidual , Peptídeos/química , Nanoestruturas/química , Nanofibras/química , Hidrogéis
17.
ChemMedChem ; 18(5): e202200654, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36604305

RESUMO

The microbiota-gut-brain axis (GBA) plays a critical role in the development of neurodegenerative diseases. Dysbiosis of the intestinal microbiome causes a significant alteration in the gut microbiota of Alzheimer's disease (AD) patients, followed by neuroinflammatory processes. Thus, AD beginning in the gut is closely related to an imbalance in gut microbiota, and hence a multidomain approach to reduce this imbalance by exerting positive effects on the gut microbiota is needed. In one example, a tyrosine-based short peptide amphiphile (sPA) was used to synthesize antibacterial AgNPs-sPA nanostructures. Such nanostructures showed high biocompatibility and low cytotoxicity, and therefore work as model drug delivery agents for addressing local bacterial infections. These may have therapeutic value for the treatment of microbiota-triggered progression of neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Infecções Bacterianas , Nanopartículas , Nanoestruturas , Doenças Neurodegenerativas , Humanos , Prata , Peptídeos , Encéfalo
18.
Mol Pharm ; 20(2): 1256-1268, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36648435

RESUMO

Four peptide amphiphiles (PA1-4) with different degrees of polymerization (DP = 40, 15, 10, and 6) were synthesized by Fuchs-Farthing and ring-opening polymerization followed by post-polymerization modification, as fully characterized by 1H NMR, FT-IR, gel permeation chromatography, and circular dichroism (CD) spectroscopy. It was found that PAs could self-assemble to form regular spherical micelles in low-concentration (about 1 mg/mL) aqueous solution, which had different contents of secondary structures and mainly adopted random coil conformations. The water solubility of PAs increases with the increase of DP, the polypeptide chain stretches randomly in water, the ß-sheets decrease, and the random coil conformations dominate. When the pH of PA solution decreases or increases, intramolecular hydrogen bonds break, and molecular chains stretch, leading to a decrease of α-helix, turn conformations, and an increase of ß-sheets. Meanwhile, the particle size of micelles increases. At around 0.4 mg/mL, the hemolysis ability of PA2 is negligible at pH 7.4 and 6.5 and about 33% at pH 5.5. Cisplatin (CDDP) was linked to micelles by coordination bonds to explore their potential as drug carriers, exhibiting controlled pH and reduction in dual drug release effects. MTT assay showed that the HeLa cell viability was 78% when cultured in the 13.5 µg/mL PA2 blank micelles for 2 days, while the cell viability was 60% in the CDDP-loaded micelles. Furthermore, a high concentration of PA2 (about 100 mg/mL) could self-assemble into a fibrous hydrogel at pH 5.5, which self-healed 2 h after incision and self-degraded 71% within 14 days. The CDDP-loaded fiber hydrogel exhibited a sustained release effect similar to the CDDP-loaded micelles. The cytotoxicity of CDDP-loaded fibers at 48 h was detected to be the same as that of the same amount of CDDP, and the cell viability was 7%. Therefore, we provide a new strategy for the synthesis of amphiphilic peptides with potential applications in nano-drug carriers and cancer therapy.


Assuntos
Cisteína , Micelas , Humanos , Células HeLa , Polimerização , Liberação Controlada de Fármacos , Espectroscopia de Infravermelho com Transformada de Fourier , Peptídeos/química , Portadores de Fármacos/química , Cisplatino , Água/química , Concentração de Íons de Hidrogênio
19.
Cell Stem Cell ; 30(2): 219-238.e14, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36638801

RESUMO

Human induced pluripotent stem cell (hiPSC) technologies offer a unique resource for modeling neurological diseases. However, iPSC models are fraught with technical limitations including abnormal aggregation and inefficient maturation of differentiated neurons. These problems are in part due to the absence of synergistic cues of the native extracellular matrix (ECM). We report on the use of three artificial ECMs based on peptide amphiphile (PA) supramolecular nanofibers. All nanofibers display the laminin-derived IKVAV signal on their surface but differ in the nature of their non-bioactive domains. We find that nanofibers with greater intensity of internal supramolecular motion have enhanced bioactivity toward hiPSC-derived motor and cortical neurons. Proteomic, biochemical, and functional assays reveal that highly mobile PA scaffolds caused enhanced ß1-integrin pathway activation, reduced aggregation, increased arborization, and matured electrophysiological activity of neurons. Our work highlights the importance of designing biomimetic ECMs to study the development, function, and dysfunction of human neurons.


Assuntos
Células-Tronco Pluripotentes Induzidas , Nanofibras , Humanos , Proteômica , Neurônios/metabolismo , Matriz Extracelular/metabolismo , Nanofibras/química
20.
Matter ; 6(2): 583-604, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36531610

RESUMO

Coronaviruses have historically precipitated global pandemics of severe acute respiratory syndrome (SARS) into devastating public health crises. Despite the virus's rapid rate of mutation, all SARS coronavirus 2 (SARS-CoV-2) variants are known to gain entry into host cells primarily through complexation with angiotensin-converting enzyme 2 (ACE2). Although ACE2 has potential as a druggable decoy to block viral entry, its clinical use is complicated by its essential biological role as a carboxypeptidase and hindered by its structural and chemical instability. Here we designed supramolecular filaments, called fACE2, that can silence ACE2's enzymatic activity and immobilize ACE2 to their surface through enzyme-substrate complexation. This docking strategy enables ACE2 to be effectively delivered in inhalable aerosols and improves its structural stability and functional preservation. fACE2 exhibits enhanced and prolonged inhibition of viral entry compared with ACE2 alone while mitigating lung injury in vivo.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA