Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 464
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 171(1): 10-13, 2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28888322

RESUMO

Michael N. Hall is this year's recipient of the Lasker Basic Medical Research Award for the identification of the target of rapamycin, TOR. TOR is a master regulator of the cell's growth and metabolic state, and its dysregulation contributes to a variety of diseases, including diabetes, obesity, neurodegenerative disorders, aging, and cancer, making the TOR pathway an attractive therapeutic target.


Assuntos
Distinções e Prêmios , Células/metabolismo , Fisiologia/história , Transdução de Sinais , Serina-Treonina Quinases TOR/fisiologia , Animais , Antifúngicos/química , Antifúngicos/isolamento & purificação , Antifúngicos/uso terapêutico , História do Século XX , Humanos , Neoplasias/tratamento farmacológico , Sirolimo/química , Sirolimo/isolamento & purificação , Sirolimo/uso terapêutico , Suíça
2.
Nano Lett ; 24(17): 5214-5223, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38649327

RESUMO

Stroke is a leading cause of global mortality and severe disability. However, current strategies used for treating ischemic stroke lack specific targeting capabilities, exhibit poor immune escape ability, and have limited drug release control. Herein, we developed an ROS-responsive nanocarrier for targeted delivery of the neuroprotective agent rapamycin (RAPA) to mitigate ischemic brain damage. The nanocarrier consisted of a sulfated chitosan (SCS) polymer core modified with a ROS-responsive boronic ester enveloped by a red blood cell membrane shell incorporating a stroke homing peptide. When encountering high levels of intracellular ROS in ischemic brain tissues, the release of SCS combined with RAPA from nanoparticle disintegration facilitates effective microglia polarization and, in turn, maintains blood-brain barrier integrity, reduces cerebral infarction, and promotes cerebral neurovascular remodeling in a mouse stroke model involving transient middle cerebral artery occlusion (tMCAO). This work offers a promising strategy to treat ischemic stroke therapy.


Assuntos
Barreira Hematoencefálica , Quitosana , Portadores de Fármacos , AVC Isquêmico , Nanopartículas , Sirolimo , Animais , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , Camundongos , Quitosana/química , Portadores de Fármacos/química , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Sirolimo/farmacologia , Sirolimo/química , Sirolimo/uso terapêutico , Nanopartículas/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Polissacarídeos/química , Polissacarídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sulfatos/química , Sulfatos/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo
3.
Small ; 20(43): e2402141, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38953313

RESUMO

Abdominal aortic aneurysm (AAA) represents a critical cardiovascular condition characterized by localized dilation of the abdominal aorta, carrying a significant risk of rupture and mortality. Current treatment options are limited, necessitating novel therapeutic approaches. This study investigates the potential of a pioneering nanodrug delivery system, RAP@PFB, in mitigating AAA progression. RAP@PFB integrates pentagalloyl glucose (PGG) and rapamycin (RAP) within a metal-organic-framework (MOF) structure through a facile assembly process, ensuring remarkable drug loading capacity and colloidal stability. The synergistic effects of PGG, a polyphenolic antioxidant, and RAP, an mTOR inhibitor, collectively regulate key players in AAA pathogenesis, such as macrophages and smooth muscle cells (SMCs). In macrophages, RAP@PFB efficiently scavenges various free radicals, suppresses inflammation, and promotes M1-to-M2 phenotype repolarization. In SMCs, it inhibits apoptosis and calcification, thereby stabilizing the extracellular matrix and reducing the risk of AAA rupture. Administered intravenously, RAP@PFB exhibits effective accumulation at the AAA site, demonstrating robust efficacy in reducing AAA progression through multiple mechanisms. Moreover, RAP@PFB demonstrates favorable biosafety profiles, supporting its potential translation into clinical applications for AAA therapy.


Assuntos
Aneurisma da Aorta Abdominal , Matriz Extracelular , Inflamação , Nanomedicina , Sirolimo , Matriz Extracelular/metabolismo , Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Nanomedicina/métodos , Inflamação/tratamento farmacológico , Inflamação/patologia , Animais , Sirolimo/farmacologia , Sirolimo/química , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Camundongos , Humanos , Sinergismo Farmacológico , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Estruturas Metalorgânicas/química , Células RAW 264.7 , Taninos Hidrolisáveis
4.
J Nanobiotechnology ; 22(1): 417, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39014353

RESUMO

Sirolimus (SR) is a macrolide with antifungal and antitumor immunosuppressant properties, classified as a selective inhibitor of mammalian target of rapamycin (mTOR). In this study, an ionic in situ gel of SR (SR-SUS-ISG) was formulated using gellan gum, exhibiting stability regardless of temperature and pH variations, causing minimal irritation. Harnessing the physiological conditions of the eye, SR-SUS-ISG underwent gelation upon contact with ions, increasing drug viscosity and prolonging retention on the ocular surface. Concurrently, SR-SUS-ISG displayed favorable shear dilution properties, reducing viscosity at ambient temperature, enhancing fluidity, and facilitating convenient packaging and transport. Biocompatibility assessments on both human corneal epithelial cells and rabbit eyes demonstrated that SR-SUS-ISG could well be tolerated. Pharmacokinetic investigations in rabbit ocular aqueous humor revealed sustained release, improved corneal penetration, and enhanced bioavailability. Additionally, in a rat corneal alkali burn model, SR-SUS-ISG exhibited inhibitory effects on corneal neovascularization, associated with decreased levels of the inflammatory factors VEGF and MMPs. These findings suggested that SR-SUS-ISG held promise as an effective ocular drug delivery system.


Assuntos
Géis , Sirolimo , Animais , Coelhos , Sirolimo/farmacologia , Sirolimo/farmacocinética , Sirolimo/química , Humanos , Géis/química , Córnea/efeitos dos fármacos , Córnea/metabolismo , Ratos , Masculino , Polissacarídeos Bacterianos/química , Nanopartículas/química , Administração Oftálmica , Neovascularização da Córnea/tratamento farmacológico , Ratos Sprague-Dawley , Viscosidade , Sistemas de Liberação de Medicamentos , Soluções Oftálmicas/química , Soluções Oftálmicas/farmacologia , Linhagem Celular , Disponibilidade Biológica
5.
Biophys J ; 122(12): 2518-2530, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37290437

RESUMO

Single-molecule data are of great significance in biology, chemistry, and medicine. However, new experimental tools to characterize, in a multiplexed manner, protein bond rupture under force are still needed. Acoustic force spectroscopy is an emerging manipulation technique which generates acoustic waves to apply force in parallel on multiple microbeads tethered to a surface. We here exploit this configuration in combination with the recently developed modular junctured-DNA scaffold that has been designed to study protein-protein interactions at the single-molecule level. By applying repetitive constant force steps on the FKBP12-rapamycin-FRB complex, we measure its unbinding kinetics under force at the single-bond level. Special efforts are made in analyzing the data to identify potential pitfalls. We propose a calibration method allowing in situ force determination during the course of the unbinding measurement. We compare our results with well-established techniques, such as magnetic tweezers, to ensure their accuracy. We also apply our strategy to study the force-dependent rupture of a single-domain antibody with its antigen. Overall, we get a good agreement with the published parameters that have been obtained at zero force and population level. Thus, our technique offers single-molecule precision for multiplexed measurements of interactions of biotechnological and medical interest.


Assuntos
Acústica , DNA , Proteínas , Análise Espectral , Análise Espectral/métodos , DNA/química , Proteínas/química , Mapas de Interação de Proteínas , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/metabolismo , Sirolimo/química , Sirolimo/metabolismo , Proteína 1A de Ligação a Tacrolimo/química , Proteína 1A de Ligação a Tacrolimo/metabolismo
6.
Nat Methods ; 17(9): 928-936, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32747768

RESUMO

Chemically inducible dimerization (CID) uses a small molecule to induce binding of two different proteins. CID tools such as the FK506-binding protein-FKBP-rapamycin-binding- (FKBP-FRB)-rapamycin system have been widely used to probe molecular events inside and outside cells. While various CID tools are available, chemically inducible trimerization (CIT) does not exist, due to inherent challenges in designing a chemical that simultaneously binds three proteins with high affinity and specificity. Here, we developed CIT by rationally splitting FRB and FKBP. Cellular and structural datasets showed efficient trimerization of split pairs of FRB or FKBP with full-length FKBP or FRB, respectively, by rapamycin. CIT rapidly induced tri-organellar junctions and perturbed intended membrane lipids exclusively at select membrane contact sites. By conferring one additional condition to what is achievable with CID, CIT expands the types of manipulation in single live cells to address cell biology questions otherwise intractable and engineer cell functions for future synthetic biology applications.


Assuntos
Sirolimo/química , Serina-Treonina Quinases TOR/química , Proteínas de Ligação a Tacrolimo/química , Células HeLa , Humanos , Modelos Moleculares , Mutação , Conformação Proteica
7.
Nat Chem Biol ; 17(12): 1262-1270, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34663942

RESUMO

DNA deaminase enzymes play key roles in immunity and have recently been harnessed for their biotechnological applications. In base editors (BEs), the combination of DNA deaminase mutator activity with CRISPR-Cas localization confers the powerful ability to directly convert one target DNA base into another. While efforts have been made to improve targeting efficiency and precision, all BEs so far use a constitutively active DNA deaminase. The absence of regulatory control over promiscuous deaminase activity remains a major limitation to accessing the widespread potential of BEs. Here, we reveal sites that permit splitting of DNA cytosine deaminases into two inactive fragments, whose reapproximation reconstitutes activity. These findings allow for the development of split-engineered BEs (seBEs), which newly enable small-molecule control over targeted mutator activity. We show that the seBE strategy facilitates robust regulated editing with BE scaffolds containing diverse deaminases, offering a generalizable solution for temporally controlling precision genome editing.


Assuntos
Nucleosídeo Desaminases/química , Biotecnologia , Sistemas CRISPR-Cas , Citosina/química , DNA/química , Quebras de DNA de Cadeia Dupla , Escherichia coli , Edição de Genes , Conformação de Ácido Nucleico , Nucleosídeo Desaminases/genética , Sirolimo/química
8.
Proc Natl Acad Sci U S A ; 117(29): 17195-17203, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32606248

RESUMO

The vast majority of intracellular protein targets are refractory toward small-molecule therapeutic engagement, and additional therapeutic modalities are needed to overcome this deficiency. Here, the identification and characterization of a natural product, WDB002, reveals a therapeutic modality that dramatically expands the currently accepted limits of druggability. WDB002, in complex with the FK506-binding protein (FKBP12), potently and selectively binds the human centrosomal protein 250 (CEP250), resulting in disruption of CEP250 function in cells. The recognition mode is unprecedented in that the targeted domain of CEP250 is a coiled coil and is topologically featureless, embodying both a structural motif and surface topology previously considered on the extreme limits of "undruggability" for an intracellular target. Structural studies reveal extensive protein-WDB002 and protein-protein contacts, with the latter being distinct from those seen in FKBP12 ternary complexes formed by FK506 and rapamycin. Outward-facing structural changes in a bound small molecule can thus reprogram FKBP12 to engage diverse, otherwise "undruggable" targets. The flat-targeting modality demonstrated here has the potential to expand the druggable target range of small-molecule therapeutics. As CEP250 was recently found to be an interaction partner with the Nsp13 protein of the SARS-CoV-2 virus that causes COVID-19 disease, it is possible that WDB002 or an analog may exert useful antiviral activity through its ability to form high-affinity ternary complexes containing CEP250 and FKBP12.


Assuntos
Actinobacteria/genética , Antivirais/farmacologia , Genoma Bacteriano , Macrolídeos/farmacologia , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína 1A de Ligação a Tacrolimo/química , Proteína 1A de Ligação a Tacrolimo/metabolismo , Actinobacteria/metabolismo , Sequência de Aminoácidos , Antivirais/química , Antivirais/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Calcineurina/genética , Calcineurina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Evolução Molecular , Células HEK293 , Humanos , Macrolídeos/química , Macrolídeos/metabolismo , Modelos Moleculares , Conformação Proteica , Homologia de Sequência , Sirolimo/química , Sirolimo/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
9.
Sensors (Basel) ; 23(7)2023 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-37050557

RESUMO

Imaging protein-protein interactions (PPIs) is a hot topic in molecular medicine in the postgenomic sequencing era. In the present study, we report bright and highly sensitive single-chain molecular strain probe templates which embed full-length Renilla luciferase 8.6-535SG (RLuc86SG) or Artificial luciferase 49 (ALuc49) as reporters. These reporters were deployed between FKBP-rapamycin binding domain (FRB) and FK506-binding protein (FKBP) as a PPI model. This unique molecular design was conceptualized to exploit molecular strains of the sandwiched reporters appended by rapamycin-triggered intramolecular PPIs. The ligand-sensing properties of the templates were maximized by interface truncations and substrate modulation. The highest fold intensities, 9.4 and 16.6, of the templates were accomplished with RLuc86SG and ALuc49, respectively. The spectra of the templates, according to substrates, revealed that the colors are tunable to blue, green, and yellow. The putative substrate-binding chemistry and the working mechanisms of the probes were computationally modeled in the presence or absence of rapamycin. Considering that the molecular strain probe templates are applicable to other PPI models, the present approach would broaden the scope of the bioassay toolbox, which harnesses the privilege of luciferase reporters and the unique concept of the molecular strain probes into bioassays and molecular imaging.


Assuntos
Sondas Moleculares , Proteínas de Ligação a Tacrolimo , Ligação Proteica , Luciferases/genética , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/metabolismo , Sirolimo/química , Sirolimo/metabolismo
10.
PLoS Biol ; 17(5): e3000252, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31112550

RESUMO

Rapamycin (Rap) and its derivatives, called rapalogs, are being explored in clinical trials targeting cancer and neurodegeneration. The underlying mechanisms of Rap actions, however, are not well understood. Mechanistic target of rapamycin (mTOR), a lysosome-localized protein kinase that acts as a critical regulator of cellular growth, is believed to mediate most Rap actions. Here, we identified mucolipin 1 (transient receptor potential channel mucolipin 1 [TRPML1], also known as MCOLN1), the principle Ca2+ release channel in the lysosome, as another direct target of Rap. Patch-clamping of isolated lysosomal membranes showed that micromolar concentrations of Rap and some rapalogs activated lysosomal TRPML1 directly and specifically. Pharmacological inhibition or genetic inactivation of mTOR failed to mimic the Rap effect. In vitro binding assays revealed that Rap bound directly to purified TRPML1 proteins with a micromolar affinity. In both healthy and disease human fibroblasts, Rap and rapalogs induced autophagic flux via nuclear translocation of transcription factor EB (TFEB). However, such effects were abolished in TRPML1-deficient cells or by TRPML1 inhibitors. Hence, Rap and rapalogs promote autophagy via a TRPML1-dependent mechanism. Given the demonstrated roles of TRPML1 and TFEB in cellular clearance, we propose that lysosomal TRPML1 may contribute a significant portion to the in vivo neuroprotective and anti-aging effects of Rap via an augmentation of autophagy and lysosomal biogenesis.


Assuntos
Lisossomos/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Cálcio/farmacologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Sirolimo/análogos & derivados , Sirolimo/química
11.
Biomacromolecules ; 23(8): 3116-3129, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35786858

RESUMO

Rapalogues are powerful therapeutic modalities for breast cancer; however, they suffer from low solubility and dose-limiting side effects. To overcome these challenges, we developed a long-circulating multiheaded drug carrier called 5FA, which contains rapamycin-binding domains linked with elastin-like polypeptides (ELPs). To target these "Hydra-ELPs" toward breast cancer, we here linked 5FA with four distinct peptides which are reported to engage the cell surface form of the 78 kDa glucose-regulated protein (csGRP78). To determine if these peptides affected the carrier solubility, this library was characterized by light scattering and mass spectrometry. To guide in vitro selection of the most potent functional carrier for rapamycin, its uptake and inhibition of mTORC1 were monitored in a ductal breast cancer model (BT474). Using flow cytometry to track cellular association, it was found that only the targeted carriers enhanced cellular uptake and were susceptible to proteolysis by SubA, which specifically targets csGRP78. The functional inhibition of mTOR was monitored by Western blot for pS6K, whereby the best carrier L-5FA reduced mTOR activity by 3-fold compared to 5FA or free rapamycin. L-5FA was further visualized using super-resolution confocal laser scanning microscopy, which revealed that targeting increased exposure to the carrier by ∼8-fold. This study demonstrates how peptide ligands for GRP78, such as the L peptide (RLLDTNRPLLPY), may be incorporated into protein-based drug carriers to enhance targeting.


Assuntos
Neoplasias da Mama , Hydra , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Portadores de Fármacos/química , Elastina/química , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Hydra/metabolismo , Peptídeos/química , Sirolimo/química , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/uso terapêutico
12.
J Nanobiotechnology ; 19(1): 331, 2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34674712

RESUMO

BACKGROUND: Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic strategies that control inflammation to treat I/R are less than satisfactory. RESULTS: We report a kind of shield and sword nano-soldier functionalized nanoparticles (monocyte membranes-coated rapamycin nanoparticles, McM/RNPs) that can reduce inflammation and relieve I/R injury by blocking monocyte infiltration and inhibiting microglia proliferation. The fabricated McM/RNPs can actively target and bind to inflammatory endothelial cells, which inhibit the adhesion of monocytes to the endothelium, thus acting as a shield. Subsequently, McM/RNPs can penetrate the endothelium to reach the injury site, similar to a sword, and release the RAP drug to inhibit the proliferation of inflammatory cells. In a rat I/R injury model, McM/RNPs exhibited improved active homing to I/R injury areas and greatly ameliorated neuroscores and infarct volume. Importantly, in vivo animal studies revealed good safety for McM/RNPs treatment. CONCLUSION: The results demonstrated that the developed McM/RNPs may serve as an effective and safe nanovehicles for I/R injury therapy.


Assuntos
Membrana Celular/química , AVC Isquêmico/metabolismo , Monócitos/citologia , Nanopartículas/química , Traumatismo por Reperfusão/metabolismo , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Masculino , Sistemas de Liberação de Fármacos por Nanopartículas , Ratos , Ratos Sprague-Dawley , Sirolimo/química , Sirolimo/farmacocinética , Sirolimo/farmacologia
13.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789284

RESUMO

BACKGROUND: Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS: In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS: Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.


Assuntos
Hiperplasia/metabolismo , Macrófagos/citologia , Sistemas de Liberação de Fármacos por Nanopartículas , Espécies Reativas de Oxigênio/metabolismo , Túnica Íntima , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Membrana Celular/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistemas de Liberação de Fármacos por Nanopartículas/química , Sistemas de Liberação de Fármacos por Nanopartículas/metabolismo , Sirolimo/química , Sirolimo/farmacocinética , Sirolimo/farmacologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Peixe-Zebra
14.
Int J Mol Sci ; 22(11)2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-34073521

RESUMO

In this study, we investigated the effect of mTOR inhibitor (mTORi) drug-eluting biodegradable stent (DE stent), a putative restenosis-inhibiting device for coronary artery, on thermal-injury-related ureteral stricture in rabbits. In vitro evaluation confirmed the dose-dependent effect of mTORi, i.e., rapamycin, on fibrotic markers in ureteral component cell lines. Upper ureteral fibrosis was induced by ureteral thermal injury in open surgery, which was followed by insertion of biodegradable stents, with or without rapamycin drug-eluting. Immunohistochemistry and Western blotting were performed 4 weeks after the operation to determine gross anatomy changes, collagen deposition, expression of epithelial-mesenchymal transition markers, including Smad, α-SMA, and SNAI 1. Ureteral thermal injury resulted in severe ipsilateral hydronephrosis. The levels of type III collagen, Smad, α-SMA, and SNAI 1 were increased 28 days after ureteral thermal injury. Treatment with mTORi-eluting biodegradable stents significantly attenuated thermal injury-induced urinary tract obstruction and reduced the level of fibrosis proteins, i.e., type III collagen. TGF-ß and EMT signaling pathway markers, Smad and SNAI 1, were significantly modified in DE stent-treated thermal-injury-related ureteral stricture rabbits. These results suggested that intra-ureteral administration of rapamycin by DE stent provides modification of fibrosis signaling pathway, and inhibiting mTOR may result in fibrotic process change.


Assuntos
Implantes Absorvíveis , Stents Farmacológicos , Sirolimo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Obstrução Ureteral , Animais , Fibrose , Coelhos , Sirolimo/química , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Obstrução Ureteral/terapia
15.
AAPS PharmSciTech ; 22(3): 108, 2021 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-33718989

RESUMO

The combination of self-microemulsifying drug delivery system (SMEDDS) and mesoporous silica materials favors the oral delivery of poorly water-soluble drugs (PWSD). However, the influence of the surface property of the mesopores towards the drug release and in vivo pharmacokinetics is still unknown. In this study, SBA-15 with hydroxyl groups (SBA-15-H), methyl groups (SBA-15-M), amino groups (SBA-15-A), or carboxyl groups (SBA-15-C) was combined with SMEDDS containing sirolimus (SRL). The diffusion and self-emulsifying of SMEDDS greatly improved the drug release over the raw SRL and SRL-SBA-15-R (R referred to as the functional groups). Results of drug absorption and X-ray photoelectron spectroscopy (XPS) showed strong hydrogen binding between SRL and the amino groups of SBA-15-A, which hindered the drug release and oral bioavailability of SRL-SMEDDS-SBA-15-A. The favorable release of SRL-SMEDDS-SBA-15-C (91.31 ± 0.57%) and SRL-SMEDDS-SBA-15-M (91.76 ± 3.72%) contributed to enhancing the maximum blood concentration (Cmax) and the area under the concentration-time curve (AUC0→48). In conclusion, the release of SRL-SMEDDS-SBA-15-R was determined by the surface affinity of the SBA-15-R and the interaction between the SRL molecules and the surface of SBA-15-R. This study suggested that the SMEDDS-SBA-15 was a favorable carrier for PWSD, and the surface property of the mesopores should be considered for the optimization of the SMEDDS-SBA-15.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos/fisiologia , Absorção Intestinal/fisiologia , Sirolimo/administração & dosagem , Sirolimo/farmacocinética , Administração Oral , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Antibacterianos/farmacocinética , Disponibilidade Biológica , Cães , Emulsões/administração & dosagem , Emulsões/química , Emulsões/farmacocinética , Absorção Intestinal/efeitos dos fármacos , Masculino , Dióxido de Silício/administração & dosagem , Dióxido de Silício/química , Dióxido de Silício/farmacocinética , Sirolimo/química , Solubilidade , Propriedades de Superfície
16.
Bioorg Med Chem Lett ; 30(11): 127134, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32253062

RESUMO

The rate of drug-induced proliferation (DIP) has been proposed as an unbiased alternative drug effect metric. However, current assays are not easy and precise enough to track minor changes in cell growth. Here, we report the optimized EZMTT based detection method which can continuously measure time-dependent growth after drug treatment and reliably detect partial drug resistance for cancer cells. Importantly, tracking time-dependent growth after drug treatment demonstrated that a KGA allosteric inhibitor alone failed to completely inhibit cancer cell growth, but a drug combination was able to provide complete inhibition in cell-based assays that translated well in in vivo animal experiments. In conclusion, this simple EZMTT method provided precise measurement of loss of susceptibility after drug treatment and has great potential to be developed for drug efficacy and drug combination studies to solve the unmet medical need.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Regulação Alostérica/efeitos dos fármacos , Antineoplásicos/química , Linhagem Celular Tumoral , Sinergismo Farmacológico , Glutaminase/química , Glutaminase/metabolismo , Humanos , Concentração Inibidora 50 , Paclitaxel/química , Paclitaxel/farmacologia , Sirolimo/química , Sirolimo/farmacologia
17.
J Cell Physiol ; 234(4): 4702-4712, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30191977

RESUMO

Chemical burns are a major cause of corneal haze and blindness. Corticosteroids are commonly used after corneal burns to attenuate the severity of the inflammation-related fibrosis. While research efforts have been aimed toward application of novel therapeutics. In the current study, a novel drug delivery system based nanostructured lipid carriers (NLCs) were designed to treat corneal alkaline burn injury. Rapamycin, a potent inhibitor of mammalian target of rapamycin pathway, was loaded in NLCs (rapa-NLCs), and the NLCs were characterized. Cell viability assay, cellular uptake of NLCs, and in vitro evaluation of the fibrotic/angiogenic genes suppression by rapa-NLCs were carried out on human isolated corneal fibroblasts. Immunohistochemistry (IHC) assays were also performed after treatment of murine model of corneal alkaline burn with rapa-NLCs. According to the results, rapamycin was efficiently loaded in NLCs. NLCs could enhance coumarin-6 fibroblast uptake by 1.5 times. Rapa-NLCs efficiently downregulated platelet-derived growth factor and transforming growth factor beta genes in vitro. Furthermore, proliferation of fibroblasts, a major cause of corneal haze after injury, reduced. IHC staining of treated cornea with alpha-smooth muscle actin and CD34 + antibodies showed efficient prevention of myofibroblasts differentiation and angiogenesis, respectively. In conclusion, ocular delivery of rapamycin using NLCs after corneal injury may be considered as a promising antifibrotic/angiogenic treatment approach to preserve patient eyesight.


Assuntos
Queimaduras Químicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Córnea/efeitos dos fármacos , Lesões da Córnea/tratamento farmacológico , Opacidade da Córnea/tratamento farmacológico , Portadores de Fármacos , Queimaduras Oculares/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Lipídeos/química , Nanopartículas , Sirolimo/administração & dosagem , Administração Oftálmica , Animais , Queimaduras Químicas/etiologia , Queimaduras Químicas/metabolismo , Queimaduras Químicas/patologia , Células Cultivadas , Córnea/metabolismo , Córnea/patologia , Lesões da Córnea/induzido quimicamente , Lesões da Córnea/metabolismo , Lesões da Córnea/patologia , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Neovascularização da Córnea/prevenção & controle , Opacidade da Córnea/induzido quimicamente , Opacidade da Córnea/metabolismo , Opacidade da Córnea/patologia , Modelos Animais de Doenças , Composição de Medicamentos , Queimaduras Oculares/induzido quimicamente , Queimaduras Oculares/metabolismo , Queimaduras Oculares/patologia , Fibroblastos/metabolismo , Fibrose , Humanos , Masculino , Camundongos Endogâmicos BALB C , Nanomedicina , Sirolimo/química , Hidróxido de Sódio , Cicatrização/efeitos dos fármacos
18.
Anal Chem ; 91(24): 15982-15987, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31738054

RESUMO

Fluorescent nanoprobes based on peptide-functionalized gold nanoparticles (AuNPs) have been widely used in bioassays. The Au-Se bond is considered as a better candidate than the Au-S bond to link the peptides and AuNPs due to the stronger ability against interference of intracellular thiol. However, the current synthetic methods for preparing peptide/AuNPs nanoprobes are always complex and time-consuming. Developing a convenient and rapid method to synthesize the Au-Se bond based nanoprobes is expected to further facilitate their application in fundamental research. Herein, we present a facile and rapid approach to prepare the Au-Se-peptide nanoprobes through a direct freezing process, which is easy-to-operate, time-saving, and surfactant-free. Compared with the traditional method, the amount of peptide loaded on AuNPs by freezing method is also promoted with 20-30%. Furthermore, the obtained nanoprobe was successfully applied to identify autophagy and apoptosis in chemotherapeutic drug treated cancer cells.


Assuntos
Congelamento , Ouro/química , Nanoestruturas/química , Peptídeos/química , Compostos de Sulfidrila/química , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Nanopartículas Metálicas/química , Microscopia Confocal , Nanoestruturas/toxicidade , Sirolimo/química , Sirolimo/farmacologia
19.
Bioconjug Chem ; 30(9): 2358-2372, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31408605

RESUMO

Elastin-Like Polypeptides (ELP) are environmentally responsive protein polymers which are easy to engineer and biocompatible, making them ideal candidates as drug carriers. Our team has recently utilized ELPs fused to FKBP12 to carry Rapamycin (Rapa), a potent immunosuppressant. Through high affinity binding to Rapa, FKBP carriers can yield beneficial therapeutic effects and reduce the off-site toxicity of Rapa. Since ICAM-1 is significantly elevated at sites of inflammation in diverse diseases, we hypothesized that a molecularly targeted ELP carrier capable of binding ICAM-1 might have advantageous properties. Here we report on the design, characterization, pharmacokinetics, and biodistribution of a new ICAM-1-targeted ELP Rapa carrier (IBPAF) and its preliminary characterization in a murine model exhibiting elevated ICAM-1. Lacrimal glands (LG) of male NOD mice, a disease model recapitulating the autoimmune dacryoadenitis seen in Sjögren's Syndrome patients, were analyzed to confirm that ICAM-1 was significantly elevated in the LG relative to control male BALB/c mice (3.5-fold, p < 0.05, n = 6). In vitro studies showed that IBPAF had significantly higher binding to TNF-α-stimulated bEnd.3 cells which overexpress surface ICAM-1, relative to nontargeted control ELP (AF)(4.0-fold, p < 0.05). A pharmacokinetics study in male NOD mice showed no significant differences between AF and IBPAF for plasma half-life, clearance, and volume of distribution. However, both constructs maintained a higher level of Rapa in systemic circulation compared to free Rapa. Interestingly, in the male NOD mouse, the accumulation of IBPAF was significantly higher in homogenized LG extracts compared to AF at 2 h (8.6 ± 6.6% versus 1.3 ± 1.3%, respectively, n = 5, p < 0.05). This accumulation was transient with no differences detected at 8 or 24 h. This study describes the first ICAM-1 targeted protein-polymer carrier for Rapa that specifically binds to ICAM-1 in vitro and accumulates in ICAM-1 overexpressing tissue in vivo, which may be useful for molecular targeting in diverse inflammatory diseases where ICAM-1 is elevated.


Assuntos
Elastina/química , Imunossupressores/química , Imunossupressores/farmacologia , Terapia de Alvo Molecular , Peptídeos/química , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Imunossupressores/metabolismo , Imunossupressores/farmacocinética , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Peptídeos/metabolismo , Peptídeos/farmacocinética , Transporte Proteico , Sirolimo/química , Distribuição Tecidual , Fator de Necrose Tumoral alfa/farmacologia
20.
BMC Cancer ; 19(1): 837, 2019 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-31455317

RESUMO

BACKGROUND: The activity of MP1, a pyrrolomycin, was studied in MYCN amplified neuroblastoma (NB) alone and combined with temsirolimus (TEM). METHODS: Activity of MP1 was tested in MYCN amplified (BE-2c, IMR) and non amplified (SKN-AS) NB cells. The effect of MP1 on MYCN, MCL-1, cleaved PARP, LC3II/LC3I, bcl-2, BAX, and BRD-4 were determined by western blot and RNAseq. The effect of MP1 on metabolism, mitochondrial morphology, and cell cycle was determined. Toxicology and efficacy of MP1 plus TEM were evaluated. RESULTS: The IC50 of MP1 was 0.096 µM in BE-2c cells compared to 0.89 µM in IMR, and >50 µM in SKN-AS. The IC50 of MP1 plus TEM in BE-2c cells was 0.023 µM. MP1 inhibited metabolism leading to quiescence and produced a decline in cell cycle S-phase. Electron microscopy showed cristae loss and rounding up of mitochondria. Gene and protein expression for MYCN and MCL-1 declined while LCII and cleaved PARP increased. Protein expression of BAX, bcl-2, and BRD-4 were not significantly changed after MP1 treatment. The in-vivo concentrations of MP1 in blood and tumor were sufficient to produce the biologic effects seen in-vitro. MP1 plus TEM produced a complete response in 3 out of 5 tumor bearing mice. In a second mouse study, the combination of MP1 and TEM slowed tumor growth compared to control. CONCLUSIONS: MP1 has a potent inhibitory effect on the viability of MYCN amplified NB. Inhibition of metabolism by MP1 induced quiescence and autophagy with a favorable toxicology and drug distribution profile. When combined with TEM anti-tumor activity was potentiated in-vitro and in-vivo.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Amplificação de Genes , Proteína Proto-Oncogênica N-Myc/genética , Pirróis/farmacologia , Sirolimo/análogos & derivados , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Interações Medicamentosas , Humanos , Camundongos , Estrutura Molecular , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/ultraestrutura , Pirróis/química , Sirolimo/química , Sirolimo/farmacologia , Análise Espectral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA