Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Nanobiotechnology ; 22(1): 300, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816719

RESUMO

BACKGROUND: Extracellular vesicles (EVs) derived from human adipose-derived mesenchymal stem cells (hADSCs) have shown great therapeutic potential in plastic and reconstructive surgery. However, the limited production and functional molecule loading of EVs hinder their clinical translation. Traditional two-dimensional culture of hADSCs results in stemness loss and cellular senescence, which is unfavorable for the production and functional molecule loading of EVs. Recent advances in regenerative medicine advocate for the use of three-dimensional culture of hADSCs to produce EVs, as it more accurately simulates their physiological state. Moreover, the successful application of EVs in tissue engineering relies on the targeted delivery of EVs to cells within biomaterial scaffolds. METHODS AND RESULTS: The hADSCs spheroids and hADSCs gelatin methacrylate (GelMA) microspheres are utilized to produce three-dimensional cultured EVs, corresponding to hADSCs spheroids-EVs and hADSCs microspheres-EVs respectively. hADSCs spheroids-EVs demonstrate excellent production and functional molecule loading compared with hADSCs microspheres-EVs. The upregulation of eight miRNAs (i.e. hsa-miR-486-5p, hsa-miR-423-5p, hsa-miR-92a-3p, hsa-miR-122-5p, hsa-miR-223-3p, hsa-miR-320a, hsa-miR-126-3p, and hsa-miR-25-3p) and the downregulation of hsa-miR-146b-5p within hADSCs spheroids-EVs show the potential of improving the fate of remaining ear chondrocytes and promoting cartilage formation probably through integrated regulatory mechanisms. Additionally, a quick and innovative pipeline is developed for isolating chondrocyte homing peptide-modified EVs (CHP-EVs) from three-dimensional dynamic cultures of hADSCs spheroids. CHP-EVs are produced by genetically fusing a CHP at the N-terminus of the exosomal surface protein LAMP2B. The CHP + LAMP2B-transfected hADSCs spheroids were cultured with wave motion to promote the secretion of CHP-EVs. A harvesting method is used to enable the time-dependent collection of CHP-EVs. The pipeline is easy to set up and quick to use for the isolation of CHP-EVs. Compared with nontagged EVs, CHP-EVs penetrate the biomaterial scaffolds and specifically deliver the therapeutic miRNAs to the remaining ear chondrocytes. Functionally, CHP-EVs show a major effect on promoting cell proliferation, reducing cell apoptosis and enhancing cartilage formation in remaining ear chondrocytes in the M1 macrophage-infiltrated microenvironment. CONCLUSIONS: In summary, an innovative pipeline is developed to obtain CHP-EVs from three-dimensional dynamic culture of hADSCs spheroids. This pipeline can be customized to increase EVs production and functional molecule loading, which meets the requirements for regulating remaining ear chondrocyte fate in the M1 macrophage-infiltrated microenvironment.


Assuntos
Condrócitos , Vesículas Extracelulares , Células-Tronco Mesenquimais , Peptídeos , Esferoides Celulares , Humanos , Condrócitos/metabolismo , Condrócitos/citologia , Vesículas Extracelulares/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Peptídeos/química , Peptídeos/metabolismo , MicroRNAs/metabolismo , MicroRNAs/genética , Macrófagos/metabolismo , Macrófagos/citologia , Células Cultivadas , Microesferas , Engenharia Tecidual/métodos , Técnicas de Cultura de Células em Três Dimensões/métodos , Microambiente Celular , Cartilagem da Orelha/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Diferenciação Celular
2.
Luminescence ; 39(5): e4764, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38684508

RESUMO

Ultrasensitive, selective, and non-invasive detection of fibrin in human serum is critical for disease diagnosis. So far, the development of high-performance and ultrasensitive biosensors maintains core challenges for biosensing. Herein, we designed a novel ribbon nanoprobe for ultrasensitive detection of fibrin. The probe contains gold nanoparticles (AuNPs) that can not only link with homing peptide Cys-Arg-Glu-Lys-Ala (CREKA) to recognize fibrin but also carry long DNA belts to form G-quadruplex-based DNAzyme, catalyzing the chemiluminescence of luminol-hydrogen peroxide (H2O2) reaction. Combined with the second amplification procedure of rolling circle amplification (RCA), the assay exhibits excellent sensitivity with a detection limit of 0.04 fmol L-1 fibrin based on the 3-sigma. Furthermore, the biosensor shows high specificity on fibrin in samples because the structure of antibody-fibrin-homing peptide was employed to double recognize fibrin. Altogether, the simple and inexpensive approach may present a great potential for reliable detection of biomarkers.


Assuntos
Técnicas Biossensoriais , Fibrina , Ouro , Nanopartículas Metálicas , Ouro/química , Nanopartículas Metálicas/química , Fibrina/química , Fibrina/análise , Humanos , DNA Catalítico/química , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/análise , Limite de Detecção , Luminol/química , Quadruplex G
3.
Molecules ; 29(11)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38893510

RESUMO

Cancer cells have higher heat sensitivity compared to normal cells; therefore, hyperthermia is a promising approach for cancer therapy because of its ability to selectively kill cancer cells by heating them. However, the specific and rapid heating of tumor tissues remains challenging. This study investigated the potential of magnetic nanoparticles (MNPs) modified with tumor-homing peptides (THPs), specifically PL1 and PL3, for tumor-specific magnetic hyperthermia therapy. The synthesis of THP-modified MNPs involved the attachment of PL1 and PL3 peptides to the surface of the MNPs, which facilitated enhanced tumor cell binding and internalization. Cell specificity studies revealed an increased uptake of PL1- and PL3-MNPs by tumor cells compared to unmodified MNPs, indicating their potential for targeted delivery. In vitro hyperthermia experiments demonstrated the efficacy of PL3-MNPs in inducing tumor cell death when exposed to an alternating magnetic field (AMF). Even without exposure to an AMF, an additional ferroptotic pathway was suggested to be mediated by the nanoparticles. Thus, this study suggests that THP-modified MNPs, particularly PL3-MNPs, hold promise as a targeted approach for tumor-specific magnetic hyperthermia therapy.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Peptídeos , Hipertermia Induzida/métodos , Humanos , Nanopartículas de Magnetita/química , Peptídeos/química , Peptídeos/farmacologia , Linhagem Celular Tumoral , Neoplasias/terapia , Neoplasias/patologia , Campos Magnéticos
4.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-37373494

RESUMO

One of the major challenges in cancer therapy lies in the limited targeting specificity exhibited by existing anti-cancer drugs. Tumor-homing peptides (THPs) have emerged as a promising solution to this issue, due to their capability to specifically bind to and accumulate in tumor tissues while minimally impacting healthy tissues. THPs are short oligopeptides that offer a superior biological safety profile, with minimal antigenicity, and faster incorporation rates into target cells/tissues. However, identifying THPs experimentally, using methods such as phage display or in vivo screening, is a complex, time-consuming task, hence the need for computational methods. In this study, we proposed StackTHPred, a novel machine learning-based framework that predicts THPs using optimal features and a stacking architecture. With an effective feature selection algorithm and three tree-based machine learning algorithms, StackTHPred has demonstrated advanced performance, surpassing existing THP prediction methods. It achieved an accuracy of 0.915 and a 0.831 Matthews Correlation Coefficient (MCC) score on the main dataset, and an accuracy of 0.883 and a 0.767 MCC score on the small dataset. StackTHPred also offers favorable interpretability, enabling researchers to better understand the intrinsic characteristics of THPs. Overall, StackTHPred is beneficial for both the exploration and identification of THPs and facilitates the development of innovative cancer therapies.


Assuntos
Neoplasias , Peptídeos , Humanos , Peptídeos/metabolismo , Oligopeptídeos , Algoritmos , Aprendizado de Máquina
5.
J Nanobiotechnology ; 20(1): 435, 2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36195937

RESUMO

Exosomes of human cardiosphere-derived cells (CDCs) are very promising for treating cardiovascular disorders. However, the current challenge is inconvenient delivery methods of exosomes for clinical application. The present study aims to explore the potential to enhance the therapeutic effect of exosome (EXO) from human CDCs to myocardial hypertrophy. A heart homing peptide (HHP) was displayed on the surface of exosomes derived from CDCs that were forced to express the HHP fused on the N-terminus of the lysosomal-associated membrane protein 2b (LAMP2b). The cardiomyocyte-targeting capability of exosomes were analyzed and their therapeutic effects were evaluated in a mouse model of myocardial hypertrophy induced by transverse aorta constriction (TAC). The molecular mechanisms of the therapeutic effects were dissected in angiotensin II-induced neonatal rat cardiomyocyte (NRCMs) hypertrophy model using a combination of biochemistry, immunohistochemistry and molecular biology techniques. We found that HHP-exosomes (HHP-EXO) accumulated more in mouse hearts after intravenous delivery and in cultured NRCMs than control exosomes (CON-EXO). Cardiac function of TAC mice was significantly improved with intravenous HHP-EXO administration. Left ventricular hypertrophy was reduced more by HHP-EXO than CON-EXO via inhibition of ß-MHC, BNP, GP130, p-STAT3, p-ERK1/2, and p-AKT. Similar results were obtained in angiotensin II-induced hypertrophy of NRCMs, in which the beneficial effects of HHP-EXO were abolished by miRNA-148a inhibition. Our results indicate that HHP-EXO preferentially target the heart and improve the therapeutic effect of CDCs-exosomes on cardiac hypertrophy. The beneficial therapeutic effect is most likely attributed to miRNA-148a-mediated suppression of GP130, which in turn inhibits STAT3/ERK1/2/AKT signaling pathway, leading to improved cardiac function and remodeling.


Assuntos
Exossomos , MicroRNAs , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Cardiomegalia/terapia , Receptor gp130 de Citocina/metabolismo , Exossomos/metabolismo , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
6.
Bioorg Med Chem Lett ; 33: 127726, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33316406

RESUMO

Fluorescence imaging is a noninvasive technique for cancer diagnosis. Dendrimers are regularly branched macromolecules with highly controllable size and structure that are a potent multifunctional nanoparticle. Anionic-terminal polyamidoamine (PAMAM) dendrimers were previously found to be accumulated in the lymph node, which is one of the main routes of tumor metastasis. In this study, we designed and synthesized a dendrimeric imaging probe for lymph node-resident tumor cell imaging. A matrix metalloproteinase-2 (MMP-2)-responsive fluorescence peptide probe and a tumor-homing peptide were conjugated to the carboxy-terminal dendrimer. The dendrimeric imaging probe treatment showed fluorescence signals inside some tumor cells (e.g., human fibrosarcoma HT-1080 and breast cancer 4T1 cells), depending on the MMP activity, but not in macrophage-like RAW264 cells.


Assuntos
Dendrímeros/farmacologia , Corantes Fluorescentes/farmacologia , Linfonodos/diagnóstico por imagem , Inibidores de Metaloproteinases de Matriz/farmacologia , Metaloproteinases da Matriz/metabolismo , Neoplasias/diagnóstico por imagem , Peptídeos/farmacologia , Animais , Linhagem Celular Tumoral , Dendrímeros/síntese química , Dendrímeros/química , Relação Dose-Resposta a Droga , Desenho de Fármacos , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Humanos , Linfonodos/metabolismo , Inibidores de Metaloproteinases de Matriz/síntese química , Inibidores de Metaloproteinases de Matriz/química , Camundongos , Estrutura Molecular , Imagem Óptica , Peptídeos/síntese química , Peptídeos/química , Relação Estrutura-Atividade
7.
Mol Ther ; 28(8): 1833-1845, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32497513

RESUMO

Systemic skin-selective therapeutics would be a major advancement in the treatment of diseases affecting the entire skin, such as recessive dystrophic epidermolysis bullosa (RDEB), which is caused by mutations in the COL7A1 gene and manifests in transforming growth factor-ß (TGF-ß)-driven fibrosis and malignant transformation. Homing peptides containing a C-terminal R/KXXR/K motif (C-end rule [CendR] sequence) activate an extravasation and tissue penetration pathway for tumor-specific drug delivery. We have previously described a homing peptide CRKDKC (CRK) that contains a cryptic CendR motif and homes to angiogenic blood vessels in wounds and tumors, but it cannot penetrate cells or tissues. In this study, we demonstrate that removal of the cysteine from CRK to expose the CendR sequence confers the peptide novel ability to home to normal skin. Fusion of the truncated CRK (tCRK) peptide to the C terminus of an extracellular matrix protein decorin (DCN), a natural TGF-ß inhibitor, resulted in a skin-homing therapeutic molecule (DCN-tCRK). Systemic DCN-tCRK administration in RDEB mice led to inhibition of TGF-ß signaling in the skin and significant improvement in the survival of RDEB mice. These results suggest that DCN-tCRK has the potential to be utilized as a novel therapeutic compound for the treatment of dermatological diseases such as RDEB.


Assuntos
Epidermólise Bolhosa/etiologia , Epidermólise Bolhosa/metabolismo , Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Animais , Biomarcadores , Modelos Animais de Doenças , Epidermólise Bolhosa/patologia , Fibrose , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Neuropilina-1/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica , Proteínas Recombinantes de Fusão/farmacologia , Pele/efeitos dos fármacos , Pele/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Cicatrização/efeitos dos fármacos
8.
Adv Exp Med Biol ; 1295: 29-48, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33543454

RESUMO

Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias/tratamento farmacológico , Peptídeos/uso terapêutico
9.
Chemistry ; 26(14): 3010-3015, 2020 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-31840306

RESUMO

Tumor-targeted drug delivery is highly important for improving chemotherapy, as it reduces the dose of cytotoxic agents and minimizes the death of healthy tissues. Towards this goal, a conjugate was synthesized of gossypol and a MCF-7 cancer cell specific CPP (cell penetrating peptide), thus providing a selective drug delivery system. Utilizing the aldehyde moiety of gossypol, the tumor homing CPP RLYMRYYSPTTRRYG was attached through a semi-labile imine linker, which was cleaved in a traceless fashion under aqueous conditions and had a half-life of approximately 10 hours. The conjugate killed MCF-7 cells to a significantly greater extent than HeLa cells or healthy fibroblasts.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Peptídeos Penetradores de Células/química , Gossipol/química , Gossipol/farmacologia , Aldeídos/química , Sequência de Aminoácidos , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Fibroblastos/citologia , Células HeLa , Humanos , Iminas/química , Células MCF-7 , Tiazolidinas/química
10.
Mol Pharm ; 17(7): 2518-2531, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32421341

RESUMO

M2-like tumor-associated macrophages (M2 TAMs) play important roles in the resistance of tumors to immunotherapies. Selective depletion or reprogramming of M2 TAMs may sensitize the nonresponsive tumors for immune-mediated eradication. However, precision delivery of payloads to M2 TAMs, while sparing healthy tissues, has remained an unresolved challenge. Here, we studied the application of a short linear peptide (CSPGAK, "mUNO") for the delivery of molecular and nanoscale cargoes in M2 TAMs in vitro and the relevance of the peptide for in vivo targeting of early-stage primary breast tumors and metastatic lung foci. First, we performed in silico modeling and found that mUNO interacts with mouse CD206 via a binding site between lectin domains CTLD1 and CTLD2, the same site previously demonstrated to be involved in mUNO binding to human CD206. Second, we showed that cultured M2 macrophages take up fluorescein-labeled (FAM) polymersomes conjugated with mUNO using the sulfhydryl group of its N-terminal cysteine. Pulse/chase studies of FAM-mUNO in M2 macrophages suggested that the peptide avoided lysosomal entrapment and escaped from early endosomes. Third, our in vivo studies with FAM-mUNO demonstrated that intraperitoneal administration results in better pharmacokinetics and higher blood bioavailability than can be achieved with intravenous administration. Intraperitoneal FAM-mUNO, but not FAM-control, showed a robust accumulation in M2-skewed macrophages in mouse models of early primary breast tumor and lung metastasis. This targeting was specific, as no uptake was observed in nonmalignant control organs, including the liver, or other cell types in the tumor, including M1 macrophages. Collectively, our studies support the application of the CD206-binding mUNO peptide for delivery of molecular and nanoscale cargoes to M2 macrophages and manifest the relevance of this mode of targeting primary and metastatic breast tumors.


Assuntos
Imunoterapia/métodos , Lectinas Tipo C/química , Neoplasias Pulmonares/diagnóstico , Metástase Linfática/diagnóstico , Lectinas de Ligação a Manose/química , Peptídeos/química , Receptores de Superfície Celular/química , Neoplasias de Mama Triplo Negativas/diagnóstico , Macrófagos Associados a Tumor/imunologia , Animais , Sítios de Ligação , Diferenciação Celular , Linhagem Celular Tumoral , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Feminino , Fluorescência , Humanos , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Metástase Linfática/diagnóstico por imagem , Metástase Linfática/imunologia , Lisossomos/metabolismo , Maleimidas/química , Receptor de Manose , Lectinas de Ligação a Manose/imunologia , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Peptídeos/administração & dosagem , Peptídeos/metabolismo , Peptídeos/farmacocinética , Poliésteres/química , Polietilenoglicóis/química , Polímeros/administração & dosagem , Polímeros/química , Polímeros/farmacologia , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/patologia , Macrófagos Associados a Tumor/metabolismo
11.
Pharm Res ; 37(4): 72, 2020 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-32215748

RESUMO

PURPOSE: Combination of PCI and chemotherapy represents a promising strategy for combating drug resistance of cancer. However, poor solubility of photosensitizers and unselectively released drugs at unwanted sites significantly impaired the treatment efficacy. Therefore, in the present study, we aimed to develop a nano-platform which could efficiently co-entrapping photosensitizers and chemotherapeutics for active targeting therapy of drug resistant cancers. METHODS: Two pro-drugs were respectively developed by covalently linking the Ce6 with each other via the GSH-sensitive linkage and the PTX with mPEG-PLA-COOH through the ROS sensitive-linker. The dual-responsive nanoparticles (PNP-Ce6) was developed by emulsion/solvent evaporation method and further modified with tLyp-1 peptides. Physicochemical properties of nanoparticles were determined by the TEM and DLC. Cellular uptake assay was investigated with the Ce6 acting as the fluorescent probe and cell growth was studied by the MTT experiment. In vivo tumor targeting and anti-tumor assay was investigated on the colorectal cancer-bearing mice. RESULTS: The developed tPNP-Ce6 were stable enough under the normal physiological conditions. However, free Ce6 and PTX were completely released when exposed the tPNP-Ce6 to the redox environment. Excellent tumor-targeting drug delivery was achieved by the tPNP-Ce6, which in turn resulted in satisfactory anit-tumor effect. Of great importance, super inhibition effect on tumor progress was achieved by the combination therapy when compared with the group only received with chemotherapy.. CONCLUSION: The results obtained in the present study indicated that the developed tPNP-Ce6 may have great potential in enhancing the therapeutic efficacy of drug-resistant colorectal cancer. Graphical Abstract Left: Targeting delivery of drug to tumor site by the tumor recognizable and dual-responsive nanoparticles and penetrating into tumor inner via the mediation of irradiation. Right: Nanoparticle distribution within tumor tissues with green represents the blood vessels stained with CD31, blue signal represents the cell nuclei stained with DAPI and red shows fluorescence of Ce6 as the indicator of the nanoparticles.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Nanopartículas , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Fotossensibilizantes/administração & dosagem , Pró-Fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cell Probes ; 51: 101530, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32035108

RESUMO

Using phage peptide library screening, we identified peptide-encoding phages that selectively home to the inflamed central nervous system (CNS) of mice with experimental autoimmune encephalomyelitis (EAE), a model of human multiple sclerosis (MS). A phage peptide display library encoding cyclic 9-amino-acid random peptides was first screened ex-vivo for binding to the CNS tissue of EAE mice, followed by in vivo screening in the diseased mice. Phage insert sequences that were present at a higher frequency in the CNS of EAE mice than in the normal (control) mice were identified by DNA sequencing. One of the phages selected in this manner, denoted as MS-1, was shown to selectively recognize CNS tissue in EAE mice. Individually cloned phages with this insert preferentially homed to EAE CNS after an intravenous injection. Similarly, systemically-administered fluorescence-labeled synthetic MS-1 peptide showed selective accumulation in the spinal cord of EAE mice. We suggest that peptide MS-1 might be useful for targeted drug delivery to CNS in EAE/MS.


Assuntos
Sistema Nervoso Central/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Peptídeos/metabolismo , Animais , Encéfalo/metabolismo , Biologia Computacional , Encefalomielite Autoimune Experimental/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Biblioteca de Peptídeos , Peptídeos/genética , Medula Espinal/metabolismo
13.
J Nanobiotechnology ; 18(1): 95, 2020 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-32660596

RESUMO

BACKGROUND: Currently, the diagnosis and treatment of neuroblastomas-the most frequent solid tumors in children-exploit the norepinephrine transporter (hNET) via radiolabeled norepinephrine analogs. We aim to develop a nanomedicine-based strategy towards precision therapy by targeting hNET cell-surface protein with hNET-derived homing peptides. RESULTS: The peptides (seq. GASNGINAYL and SLWERLAYGI) were shown to bind high-resolution homology models of hNET in silico. In particular, one unique binding site has marked the sequence and structural similarities of both peptides, while most of the contribution to the interaction was attributed to the electrostatic energy of Asn and Arg (< - 228 kJ/mol). The peptides were comprehensively characterized by computational and spectroscopic methods showing ~ 21% ß-sheets/aggregation for GASNGINAYL and ~ 27% α-helix for SLWERLAYGI. After decorating 12-nm ferritin-based nanovehicles with cysteinated peptides, both peptides exhibited high potential for use in actively targeted neuroblastoma nanotherapy with exceptional in vitro biocompatibility and stability, showing minor yet distinct influences of the peptides on the global expression profiles. Upon binding to hNET with fast binding kinetics, GASNGINAYLC peptides enabled rapid endocytosis of ferritins into neuroblastoma cells, leading to apoptosis due to increased selective cytotoxicity of transported payload ellipticine. Peptide-coated nanovehicles significantly showed higher levels of early apoptosis after 6 h than non-coated nanovehicles (11% and 7.3%, respectively). Furthermore, targeting with the GASNGINAYLC peptide led to significantly higher degree of late apoptosis compared to the SLWERLAYGIC peptide (9.3% and 4.4%, respectively). These findings were supported by increased formation of reactive oxygen species, down-regulation of survivin and Bcl-2 and up-regulated p53. CONCLUSION: This novel homing nanovehicle employing GASNGINAYLC peptide was shown to induce rapid endocytosis of ellipticine-loaded ferritins into neuroblastoma cells in selective fashion and with successful payload. Future homing peptide development via lead optimization and functional analysis can pave the way towards efficient peptide-based active delivery of nanomedicines to neuroblastoma cells.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Endocitose/genética , Nanoestruturas/química , Neuroblastoma/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ferritinas/química , Humanos , Nanomedicina , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/química , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo
14.
J Pept Sci ; 25(9): e3201, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31309656

RESUMO

Targeted delivery of antitumor drugs is especially important for tumor therapy. Cell-penetrating peptides (CPPs) have been shown to be very effective drug carriers for tumor therapy. However, most CPPs lack tumor cell specificity. Here, we identified a highly efficient CPP, CAT, from the newly identified buffalo-derived cathelicidin family, which exhibits a preferential binding capacity for multiple tumor cell lines and delivers carried drug molecules into cells. CAT showed an approximately threefold to sixfold higher translocation efficiency than some reported cell-penetrating antimicrobial peptides, including the well-known classical CPP TAT. Moreover, the delivery efficiency of CAT was greater in a variety of tested tumor cells than in normal cells, especially for the human hepatoma cell line SMMC-7721, for which delivery was 7 times more efficient than the normal human embryonic lung cell line MRC-5, according to fluorescent labeling experiment results. CAT was conjugated to the Momordica charantia-derived type-I ribosome-inactivating protein MAP 30, and the cytotoxicity of the MAP 30-CAT fusion protein in the tumor cell line SMMC-7721 was significantly enhanced compared with that of the unconjugated MAP 30. The IC50 value of MAP 30-CAT was approximately 83 times lower than the IC50 value of the original MAP 30. Interestingly, the IC50 value of MAP 30 alone for MRC-5 was approximately twofold higher than the value for SMMC-7721, showing a small difference. However, when MAP 30 was conjugated to CAT, the difference in IC50 values between the two cell lines was significantly increased by 38-fold. The results of the flow cytometric detection of apoptosis revealed that the increase in cytotoxicity after CAT conjugation was mainly caused by the increased induction of apoptosis by the fusion protein. These results suggest that CAT, as a novel tumor-homing CPP, has great potential in drug delivery applications in vivo and will be beneficial to the development of tumor therapeutics.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Antineoplásicos/farmacologia , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/isolamento & purificação , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Búfalos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Portadores de Fármacos/química , Portadores de Fármacos/isolamento & purificação , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Relação Estrutura-Atividade , Catelicidinas
15.
Bioorg Med Chem Lett ; 28(18): 3015-3024, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30031619

RESUMO

Cancer is one of the major causes of death globally. The current treatment options are insufficient, leading to unmet medical needs in cancer treatment. Off-target side effects, multidrug resistance, selective distribution to cancerous tissues, and cell membrane permeation of anti-cancer agents are critical problems to overcome. There is a method to solve these problems by using receptor-mediated endocytosis (RME). It is well known that proteins such as integrin, HER2, EGFR, or other cancer biomarkers are specifically overexpressed on the surface of target cancer cells. By taking advantage of such specific receptors, payloads can be transported into cells through endocytosis using a conjugate composed of the corresponding ligands connected to the payloads by an appropriate linker. After RME, the payloads released by endosomal escape into the cytoplasm can exhibit the cytotoxic activity against cancer cells. Cell-penetrating peptides (CPPs), tumor-homing peptides (THPs), and monoclonal antibodies (mAbs) are utilized as ligands in this system. Antibody drug conjugates (ADCs) based on RME have already been used to cure cancer. In addition to the canonical conjugate method, nanocarriers for spontaneous accumulation in cancer tissue due to enhanced permeability and retention (EPR) effect are extensively used. In this review, I introduce the possibilities and advantages of drug design and development based on RME for the treatment of cancer.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Membrana Celular/metabolismo , Sistemas de Liberação de Medicamentos , Endocitose , Neoplasias/tratamento farmacológico , Receptores de Superfície Celular/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Humanos , Neoplasias/patologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia
16.
Molecules ; 23(5)2018 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-29772690

RESUMO

Over recent decades multiple therapeutic approaches have been explored for improved management of peritoneally disseminated malignancies-a grim condition known as peritoneal carcinomatosis (PC). Intraperitoneal (IP) administration can be used to achieve elevated local concentration and extended half-life of the drugs in the peritoneal cavity to improve their anticancer efficacy. However, IP-administered chemotherapeutics have a short residence time in the IP space, and are not tumor selective. An increasing body of work suggests that functionalization of drugs and nanoparticles with targeting peptides increases their peritoneal retention and provides a robust and specific tumor binding and penetration that translates into improved therapeutic response. Here we review the progress in affinity targeting of intraperitoneal anticancer compounds, imaging agents and nanoparticles with tumor-homing peptides. We review classes of tumor-homing peptides relevant for PC targeting, payloads for peptide-guided precision delivery, applications for targeted compounds, and the effects of nanoformulation of drugs and imaging agents on affinity-based tumor delivery.


Assuntos
Peptídeos/administração & dosagem , Neoplasias Peritoneais/diagnóstico por imagem , Neoplasias Peritoneais/tratamento farmacológico , Animais , Sistemas de Liberação de Medicamentos , Humanos , Injeções Intraperitoneais , Nanopartículas , Tamanho da Partícula , Peptídeos/uso terapêutico
17.
Biochem Biophys Res Commun ; 482(4): 1042-1047, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27908724

RESUMO

The key challenge to improve the efficacy of cell therapy is how to efficiently modify cells with a specific molecule or compound that can guide the cells to the target tissue. To address this, we have developed a cell surface engineering technology to non-invasively modify the cell surface. This technology can embed a wide variety of bioactive molecules on any cell surface and allow for the targeting of a wide range of tissues in a variety of disease states. Using our cell surface engineering technology, mesenchymal stem cells (MSC)s were modified with: 1) a homing peptide or a recombinant protein to facilitate the migration of the cells toward a specific molecular target; or 2) magnetic resonance imaging (MRI) contrast agents to allow for in vivo tracking of the cells. The incorporation of a homing peptide or a targeting ligand on MSCs facilitated the migration of the cells toward their molecular target. MRI contrast agents were successfully embedded on the cell surfaces without adverse effects to the cells and the contrast agent-labeled cells were detectable by MRI. Our technology is a promising method of cell surface engineering that is applicable to a broad range of cell therapies.


Assuntos
Rastreamento de Células/métodos , Células-Tronco Mesenquimais/citologia , Linhagem Celular , Membrana Celular/química , Movimento Celular , Quimiocina CXCL12/análise , Meios de Contraste/análise , Fluoresceína-5-Isotiocianato/análise , Humanos , Ligantes , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/química , Microscopia Confocal/métodos , Peptídeos/análise , Fosfatidiletanolaminas/análise , Polietilenoglicóis/análise
18.
J Pept Sci ; 23(2): 162-171, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27862637

RESUMO

Peptides homing tumor vasculature are considered promising molecular imaging agents for cancer detection at an early stage. In addition to their high binding affinity, improved tissue penetrating ability, and low immunogenicity, they can deliver targeted anticancer drugs, thus expanding therapeutic treatments. Among those, CREKA, a linear peptide that specifically binds to clotted-plasma proteins in tumor vessels, has been recently employed to design bioactive systems able to target different cancer types. Within this context, this paper explores the biorecognition event between CR(NMe)EKA, an engineered CREKA-analog bearing a noncoded amino acid (N-methyl-Glu) that is responsible for its enhanced activity, and clotted-plasma proteins (fibrin and fibrinogen) by nanomechanical detection. Specifically, the tumor-homing peptide was covalently attached via epoxysilane chemistry onto silicon microcantilever chips that acted as sensors during dynamic mode experiments. Before that, each step of the functionalization process was followed by contact angle measurements, interferometry, X-ray photoelectron spectroscopy, and atomic force microscopy, thus revealing the applied protocol as a suitable strategy. The fibrin(ogen)-binding induced by CR(NMe)EKA was detected by the resonance frequency shift of the cantilevers, and a detection limit of 100 ng/mL was achieved for both proteins. Even though further development is required, this work reflects the promising application of emerging technologies capable of assisting in the comprehension of biological interactions and their implications in the biotechnological field. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Assuntos
Técnicas Biossensoriais/instrumentação , Fibrina/química , Fibrinogênio/química , Oligopeptídeos/química , Análise Serial de Proteínas/instrumentação , Fenômenos Biomecânicos , Técnicas Biossensoriais/métodos , Glutamatos/química , Humanos , Limite de Detecção , Oligopeptídeos/síntese química , Análise Serial de Proteínas/métodos , Ligação Proteica , Engenharia de Proteínas , Silanos/química , Silício/química , Propriedades de Superfície
19.
Biomed J ; 47(1): 100592, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37004870

RESUMO

BACKGROUND: To overcome the drawbacks of traditional therapy for corneal neovascularization (CNV), we evaluated the efficacy of polyethylene glycol (PEG)-conjugated Ala-Pro-Arg-Pro-Gly (APRPG) peptide modified dexamethasone (Dex), a novel nano-prodrug (Dex-PEG-APRPG, DPA). METHODS: Characterization of DPA nano-prodrug were measured with transmission electron microscopy (TEM) and dynamic light scattering (DLS) analyses. Cytotoxicity and effects on cell migration and tube formation of DPA were evaluated in vitro. A murine CNV model was established by cornea alkali burn. The injured corneas were given eye drops of DPA (0.2 mM), Dex solution (0.2 mM), Dexp (2 mM), or normal saline three times a day. After two weeks, eyes were obtained for the analysis of histopathology, immunostaining, and mRNA expression. RESULTS: DPA with an average diameter of 30 nm, presented little cytotoxicity and had good ocular biocompatibility. More importantly, DPA showed specific targeting to vascular endothelial cells with efficient inhibition on cell migration and tube formation. In a mouse CNV model, clinical, histological, and immunohistochemical examination results revealed DPA had a much stronger angiogenesis suppression than Dex, resembling a clinical drug with an order of magnitude higher concentration. This was ascribed to the significant downregulations in the expression of pro-angiogenic and pro-inflammatory factors in the corneas. In vivo imaging results also demonstrated that APRPG could prolong ocular retention time. CONCLUSIONS: This study suggests that DPA nano-prodrug occupies advantages of specific targeting ability and improved bioavailability over conventional therapy, and holds great potential for safe and efficient CNV therapy.


Assuntos
Neovascularização da Córnea , Pró-Fármacos , Camundongos , Animais , Neovascularização da Córnea/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Células Endoteliais , Polietilenoglicóis/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/uso terapêutico , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Neovascularização Patológica/tratamento farmacológico
20.
Stem Cell Rev Rep ; 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38814409

RESUMO

The placenta plays a crucial role in maintaining normal pregnancy. The failure of spiral artery remodeling (SAR) is a key factor leading to placental ischemia and poor perfusion which is strongly associated with obstetric diseases, including preeclampsia (PE) and fetal growth restriction (FGR). Existing interventions for PE and FGR are limited and termination of pregnancy is inevitable when the maternal or fetus condition deteriorates. Considering the safety of the mother and fetus, treatments that may penetrate the placental barrier and harm the fetus are not accepted. Developing targeted treatment strategies for these conditions is urgent and necessary. With the proven efficacy of targeted therapy in treating conditions such as endometrial cancer and trophoblastic tumors, research on placental dysfunction continues to deepen. This article reviews the studies on placenta-targeted treatment and drug delivery strategies, summarizes the characteristics proposes corresponding improvement measures in targeted treatment, provides solutions for existing problems, and makes suggestions for future studies.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa