Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 18(24): 15790-15801, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38847355

RESUMEN

Targeted drug delivery systems based on metal-organic frameworks (MOFs) have progressed tremendously since inception and are now widely applicable in diverse scientific fields. However, translating MOF agents directly to targeted drug delivery systems remains a challenge due to the biomolecular corona phenomenon. Here, we observed that supramolecular conjugation of antibodies to the surface of MOF particles (MOF-808) via electrostatic interactions and coordination bonding can reduce protein adhesion in biological environments and show stealth shields. Once antibodies are stably conjugated to particles, they were neither easily exchanged with nor covered by biomolecule proteins, which is indicative of the stealth effect. Moreover, upon conjugation of the MOF particle with specific targeted antibodies, namely, anti-CD44, human epidermal growth factor receptor 2 (HER2), and epidermal growth factor receptor (EGFR), the resulting hybrid exhibits an augmented targeting efficacy toward cancer cells overexpressing these receptors, such as HeLa, SK-BR-3, and 4T1, as evidenced by flow cytometry. The therapeutic effectiveness of the antibody-conjugated MOF (anti-M808) was further evaluated through in vivo imaging and the assessment of tumor inhibition effects using IR-780-loaded EGFR-M808 in a 4T1 tumor xenograft model employing nude mice. This study therefore provides insight into the use of supramolecular antibody conjugation as a promising method for developing MOF-based drug delivery systems.


Asunto(s)
Estructuras Metalorgánicas , Ratones Desnudos , Estructuras Metalorgánicas/química , Humanos , Animales , Ratones , Sistemas de Liberación de Medicamentos , Anticuerpos/química , Anticuerpos/inmunología , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Línea Celular Tumoral , Células HeLa , Ratones Endogámicos BALB C , Antineoplásicos/química , Antineoplásicos/farmacología , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Femenino
2.
Chem Commun (Camb) ; 60(15): 2098-2101, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38295368

RESUMEN

We report a specific lysosome targeted light-responsive CO-releasing molecule (Lyso-CORM). Lyso-CORM is very stable under dark conditions. CO and singlet oxygen (1O2) generation was effectively triggered under one photon and two photon excitation (800 nm) conditions. The cytotoxicity results demonstrated that Lyso-CORM showed good phototoxicity due to the synergistic effect of CO and 1O2 release, and its good biocompatibility, negligible dark toxicity and specific lysosome targeting make Lyso-CORM a potent candidate for phototherapeutic applications.


Asunto(s)
Compuestos Organometálicos , Oxígeno Singlete , Luz , Fotones , Lisosomas , Monóxido de Carbono , Compuestos Organometálicos/farmacología
3.
J Am Chem Soc ; 145(40): 21991-22008, 2023 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-37664981

RESUMEN

Senolytics, which eliminate senescent cells from tissues, represent an emerging therapeutic strategy for various age-related diseases. Most senolytics target antiapoptotic proteins, which are overexpressed in senescent cells, limiting specificity and inducing severe side effects. To overcome these limitations, we constructed self-assembling senolytics targeting senescent cells with an intracellular oligomerization system. Intracellular aryl-dithiol-containing peptide oligomerization occurred only inside the mitochondria of senescent cells due to selective localization of the peptides by RGD-mediated cellular uptake into integrin αvß3-overexpressed senescent cells and elevated levels of reactive oxygen species, which can be used as a chemical fuel for disulfide formation. This oligomerization results in an artificial protein-like nanoassembly with a stable α-helix secondary structure, which can disrupt the mitochondrial membrane via multivalent interactions because the mitochondrial membrane of senescent cells has weaker integrity than that of normal cells. These three specificities (integrin αvß3, high ROS, and weak mitochondrial membrane integrity) of senescent cells work in combination; therefore, this intramitochondrial oligomerization system can selectively induce apoptosis of senescent cells without side effects on normal cells. Significant reductions in key senescence markers and amelioration of retinal degeneration were observed after elimination of the senescent retinal pigment epithelium by this peptide senolytic in an age-related macular degeneration mouse model and in aged mice, and this effect was accompanied by improved visual function. This system provides a strategy for the treatment of age-related diseases using supramolecular senolytics.


Asunto(s)
Senescencia Celular , Senoterapéuticos , Ratones , Animales , Especies Reactivas de Oxígeno , Péptidos/farmacología , Integrinas
4.
J Am Chem Soc ; 145(33): 18414-18431, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37525328

RESUMEN

Lysosomes remain powerful organelles and important targets for cancer therapy because cancer cell proliferation is greatly dependent on effective lysosomal function. Recent studies have shown that lysosomal membrane permeabilization induces cell death and is an effective way to treat cancer by bypassing the classical caspase-dependent apoptotic pathway. However, most lysosome-targeted anticancer drugs have very low selectivity for cancer cells. Here, we show intra-lysosomal self-assembly of a peptide amphiphile as a powerful technique to overcome this problem. We designed a peptide amphiphile that localizes in the cancer lysosome and undergoes cathepsin B enzyme-instructed supramolecular assembly. This localized assembly induces lysosomal swelling, membrane permeabilization, and damage to the lysosome, which eventually causes caspase-independent apoptotic death of cancer cells without conventional chemotherapeutic drugs. It has specific anticancer effects and is effective against drug-resistant cancers. Moreover, this peptide amphiphile exhibits high tumor targeting when attached to a tumor-targeting ligand and causes significant inhibition of tumor growth both in cancer and drug-resistant cancer xenograft models.


Asunto(s)
Apoptosis , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Lisosomas/metabolismo , Caspasas/metabolismo , Péptidos/farmacología , Péptidos/metabolismo , Línea Celular Tumoral
5.
Small ; 19(22): e2300218, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36864579

RESUMEN

Metal-organic framework (MOF) nanoparticles have recently emerged as a promising vehicle for drug delivery with high porosity and feasibility. However, employing a MOF-based drug delivery system remains a challenge due to the difficulty in controlling interfaces of particles in a biological environment. In this paper, protein corona-blocked Zr6 -based MOF (PCN-224) nanoparticles are presented for targeted cancer therapy with high efficiency. The unmodified PCN-224 surface is precoated with glutathione transferase (GST)-fused targetable affibody (GST-Afb) proteins via simple mixing conjugations instead of chemical modifications that can induce the impairment of proteins. GST-Afb proteins are shown to stably protect the surface of PCN-224 particles in a specific orientation with GST adsorbed onto the porous surface and the GST-linked Afb posed outward, minimizing the unwanted interfacial interactions of particles with external biological proteins. The Afb-directed cell-specific targeting ability of particles and consequent induction of cell death is demonstrated both in vitro and in vivo by using two kinds of Afb, which targets the surface membrane receptor, human epidermal growth factor receptor 2 (HER2) or epidermal growth factor receptor (EGFR). This study provides insight into the way of regulating the protein-adhesive surface of MOF nanoparticles and designing a more effective MOF-hosted targeted delivery system.


Asunto(s)
Estructuras Metalorgánicas , Nanopartículas , Neoplasias , Humanos , Estructuras Metalorgánicas/metabolismo , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Proteínas de la Membrana
6.
RSC Chem Biol ; 3(12): 1416-1421, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36544576

RESUMEN

The treatment of osteosarcoma involves an adjuvant therapy that combines surgery and chemotherapy. However, considering that children are the main victims of osteosarcoma, replacing such a harsh treatment with a soft but powerful method that ensures a complete cure while having no adverse effects is highly desirable. To achieve this aim, we have developed a supramolecular therapeutic strategy based on morphology-transformable mitochondria-targeting peptides for the eradication of osteosarcoma with enhanced selectivity and reduced side effects. A newly designed micelle-forming amphiphilic peptide, l-Mito-FFYp, consisting of a phosphate substrate for the biomarker enzyme of osteosarcoma alkaline phosphatase (ALP), disassembles in response to the ALP enzyme in the cell membrane to generate positively charged l-Mito-FFY molecules, which diffuse inside the targeted cell and self-assemble to form nanostructures specifically inside the mitochondria to induce cell apoptosis.

7.
Chem Sci ; 13(21): 6197-6204, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35733910

RESUMEN

Mitochondria are essential intracellular organelles involved in many cellular processes, especially adenosine triphosphate (ATP) production. Since cancer cells require high ATP levels for proliferation, ATP elimination can be a unique target for cancer growth inhibition. We describe a newly developed mitochondria-targeting nucleopeptide (MNP) that sequesters ATP by self-assembling with ATP inside mitochondria. MNP interacts strongly with ATP through electrostatic and hydrogen bonding interactions. MNP exhibits higher binding affinity for ATP (-637.5 kJ mol-1) than for adenosine diphosphate (ADP) (-578.2 kJ mol-1). To improve anticancer efficacy, the small-sized MNP/ADP complex formed large assemblies with ATP inside cancer cell mitochondria. ATP sequestration and formation of large assemblies of the MNP/ADP-ATP complex inside mitochondria caused physical stress by large structures and metabolic disorders in cancer cells, leading to apoptosis. This work illustrates a facile approach to developing cancer therapeutics that relies on molecular assemblies.

8.
J Am Chem Soc ; 144(12): 5503-5516, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35235326

RESUMEN

Biological nanomachines, including proteins and nucleic acids whose function is activated by conformational changes, are involved in every biological process, in which their dynamic and responsive behaviors are controlled by supramolecular recognition. The development of artificial nanomachines that mimic the biological functions for potential application as therapeutics is emerging; however, it is still limited to the lower hierarchical level of the molecular components. In this work, we report a synthetic machinery nanostructure in which actuatable molecular components are integrated into a hierarchical nanomaterial in response to external stimuli to regulate biological functions. Two nanometers core-sized gold nanoparticles are covered with ligand layers as actuatable components, whose folding/unfolding motional response to the cellular environment enables the direct penetration of the nanoparticles across the cellular membrane to disrupt intracellular organelles. Furthermore, the pH-responsive conformational movements of the molecular components can induce the apoptosis of cancer cells. This strategy based on the mechanical motion of molecular components on a hierarchical nanocluster would be useful to design biomimetic nanotoxins.


Asunto(s)
Fenómenos Biológicos , Nanopartículas del Metal , Nanoestructuras , Membrana Celular , Oro , Nanoestructuras/toxicidad
9.
Chembiochem ; 22(24): 3391-3397, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34580971

RESUMEN

Cellular senescence, a stable form of cell cycle arrest, facilitates protection from tumorigenesis and aids in tissue repair as they accumulate in the body at an early age. However, long-term retention of senescent cells causes inflammation, aging of the tissue, and progression of deadly diseases such as obesity, diabetes, and atherosclerosis. Various attempts have been made to achieve selective elimination of senescent cells from the body, yet little has been explored in designing the mitochondria-targeted senolytic agent. Many characteristics of senescence are associated with mitochondria. Here we have designed a library of alkyl-monoquaternary ammonium-triphenyl phosphine (TPP) and alkyl-diquaternary ammonium-TPP of varying alkyl chain lengths, which target the mitochondria; we also studied their senolytic properties. It was observed that the alkyl-diquaternary ammonium-TPP with the longest chain length induced apoptosis in senescent cells selectively via an increase of reactive oxygen species (ROS) and mitochondrial membrane disruption. This study demonstrates that mitochondria could be a potential target for designing new small molecules as senolytic agents for the treatment of a variety of dysfunctions associated with pathological aging.


Asunto(s)
Antineoplásicos/farmacología , Membranas Mitocondriales/efectos de los fármacos , Compuestos de Amonio/química , Compuestos de Amonio/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Derivados del Benceno/química , Derivados del Benceno/farmacología , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Humanos , Ratones , Estructura Molecular , Células 3T3 NIH , Fosfinas/química , Fosfinas/farmacología
10.
ACS Nano ; 15(9): 14492-14508, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34478266

RESUMEN

Recent advances in supramolecular chemistry research have led to the development of artificial chemical systems that can form self-assembled structures that imitate proteins involved in the regulation of cellular function. However, intracellular polymerization systems that operate inside living cells have been seldom reported. In this study, we developed an intramitochondrial polymerization-induced self-assembly system for regulating the cellular fate of cancer cells. It showed that polymeric disulfide formation inside cells occurred due to the high reactive oxygen species (ROS) concentration of cancer mitochondria. This polymerization barely occurs elsewhere in the cell owing to the reductive intracellular environment. The polymerization of the thiol-containing monomers further increases the ROS level inside the mitochondria, thereby autocatalyzing the polymerization process and creating fibrous polymeric structures. This process induces dysfunction of the mitochondria, which in turn activates cell necroptosis. Thus, this in situ polymerization system shows great potential for cancer treatment, including that of drug-resistant cancers.


Asunto(s)
Disulfuros , Neoplasias , Neoplasias/tratamiento farmacológico , Polimerizacion
11.
J Phys Chem B ; 125(31): 8768-8780, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34328335

RESUMEN

The microtubule is regarded as the key target for designing anticancer and neurotherapeutic drugs due to its functional importance in eukaryotic cells including neurons. The microtubule is a dynamic hollow polymer tube consisting of α,ß-tubulin heterodimer. Polymerization of α,ß-tubulin heterodimer resulted in microtubule formation. GTP plays a crucial role in microtubule polymerization. It binds at the exchangeable binding site of the ß-tubulin heterodimer, and it is one of the most crucial therapeutic hot spots for designing anticancer therapeutics. In this manuscript, we have shown using an in silico strategy and various in vitro and cellular experiments that the binding affinity to the tubulin and cancer therapeutic potential of an exchangeable GTP/GDP binding antimitotic tetrapeptide (SP: Ser-Leu-Arg-Pro) is increased through changing proline with the multifluorine substituted proline. This study showcases the importance of the proline amino acid and its pyrrolidine ring in the regulation of binding with tubulin at the GTP binding pocket.


Asunto(s)
Antimitóticos , Tubulina (Proteína) , Antimitóticos/farmacología , Sitios de Unión , Flúor , Guanosina Trifosfato , Microtúbulos/metabolismo , Prolina , Tubulina (Proteína)/metabolismo
12.
J Mater Chem B ; 9(14): 3143-3152, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33586760

RESUMEN

Drug resistance is a major cause of treatment failure for small-molecule cancer chemotherapies, despite the advances in combination therapies, drug delivery systems, epigenetic drugs, and proteolysis-targeting chimeras. Herein, we report the use of a drug resistance-free cytotoxic nanodrug as an alternative to small-molecule drugs. The present nanodrugs comprise 2 nm core gold nanoparticles (AuNPs) covered completely with multivalent hydrocarbon chains to a final diameter of ∼10 nm as single drug molecules. This hydrophobic drug-platform was delivered in composite form (∼35 nm) with block-copolymer like other small-molecular drugs. Upon uptake by cells, the nanodrugs enhanced the intracellular levels of reactive oxygen species and induced apoptosis, presumably reflecting multivalent interactions between aliphatic chains and intracellular biomolecules. No resistance to our novel nanodrug was observed following multiple treatment passages and the potential for use in cancer therapy was verified in a breast cancer patient-derived xenograft mouse model. These findings provide insight into the use of nano-scaled compounds as agents that evade drug resistance to cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Oro/química , Oro/farmacología , Humanos , Hidrocarburos/química , Hidrocarburos/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Nanopartículas del Metal/química , Ratones , Ratones Desnudos , Tamaño de la Partícula , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
13.
Biomacromolecules ; 21(12): 4806-4813, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-32865983

RESUMEN

The intracellular or pericellular self-assembly of amphiphilic peptides is emerging as a potent cancer therapeutic strategy. Achieving the self-assembly of amphiphilic peptides inside a cell or cellular organelle is challenging due to the complex cellular environment, which consists of many amphiphilic biomolecules that may alter the self-assembling propensity of the synthetic peptides. Herein, we show that the hydrophobic-hydrophilic balance of the amphiphilic peptides determines the self-assembling propensity, thereby controlling the fate of the cell. A series of peptides were designed to target and self-assemble inside the mitochondria of cancer cells. The hydrophobicity of the peptides was tuned by varying their N-terminus capping. The analysis showed that the largest hydrophobic peptide was self-assembled before reaching the mitochondria and showed no selectivity toward cancer cells, whereas hydrophilic peptides could not self-assemble inside the mitochondria. Optimum balance between hydrophobicity and hydrophilicity is a critical factor for achieving self-assembly inside the mitochondria, thereby providing greater selectivity against cancer cells.


Asunto(s)
Neoplasias , Péptidos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Mitocondrias , Neoplasias/tratamiento farmacológico
14.
Chemistry ; 26(47): 10695-10701, 2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32428292

RESUMEN

The development of photosensitizers for cancer photodynamic therapy has been challenging due to their low photostability and therapeutic inefficacy in hypoxic tumor microenvironments. To overcome these issues, we have developed a mitochondria-targeted photosensitizer consisting of an indocyanine moiety with triphenylphosphonium arms, which can self-assemble into spherical micelles directed to mitochondria. Self-assembly of the photosensitizer resulted in a higher photostability by preventing free rotation of the indoline ring of the indocyanine moiety. The mitochondria targeting capability of the photosensitizer allowed it to utilize intramitochondrial oxygen. We found that the mitochondria-targeted photosensitizer localized to mitochondria and induced apoptosis of cancer cells both normoxic and hypoxic conditions through generation of ROS. The micellar self-assemblies of the photosensitizer were further confirmed to selectively localize to tumor tissues in a xenograft tumor mouse model through passive targeting and showed efficient tumor growth inhibition.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Ratones , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
RSC Adv ; 10(71): 43383-43388, 2020 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-35519677

RESUMEN

Controlled intracellular chemical reactions to regulate cellular functions remain a challenge in biology mimetic systems. Herein, we developed an intra-mitochondrial bio-orthogonal reaction to induce aggregation induced emission. In situ carbonyl ligation inside mitochondria drives the molecules to form nano-aggregates with green fluorescence, which leads to depolarization of the mitochondrial membrane, generation of ROS, and subsequently mitochondrial dysfunction. This intra-mitochondrial carbonyl ligation shows great potential for anticancer treatment in various cancer cell lines.

16.
Chem Commun (Camb) ; 55(84): 12631-12634, 2019 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-31580341

RESUMEN

Mitochondria targeting sensitizers are continuing to gain importance in photodynamic therapy (PDT). Members of the 90 kDa heat shock protein (Hsp90) family, including TRAP1 (Hsp75), are overexpressed in cancer cells and help to control the antiapoptotic protein activity. The present work introduces an Hsp90 inhibitor-mitochondria targeting indocyanine dye conjugate (IR-PU) for high PDT efficacy.


Asunto(s)
Antineoplásicos/química , Inhibidores Enzimáticos/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Verde de Indocianina/química , Mitocondrias/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/química , Humanos , Verde de Indocianina/farmacología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Imagen Óptica/métodos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Oxígeno Singlete/metabolismo
17.
ACS Appl Mater Interfaces ; 11(31): 27512-27520, 2019 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-31293157

RESUMEN

Metal-organic framework (MOF) nanoparticles with high porosity and greater tunability have emerged as new drug delivery vehicles. However, premature drug release still remains a challenge in the MOF delivery system. Here, we report an enzyme-responsive, polymer-coated MOF gatekeeper system using hyaluronic acid (HA) and PCN-224 nanoMOF. The external surface of nanoMOF can be stably covered by HA through multivalent coordination bonding between the Zr cluster and carboxylic acid of HA, which acts as a gatekeeper. HA allows selective accumulation of drug carriers in CD44 overexpressed cancer cells and enzyme-responsive drug release in the cancer cell environment. In particular, inherent characteristics of PCN-224, which is used as a drug carrier, facilitates the transfer of the drug to cancer cells more stably and allows photodynamic therapy. This HA-PCN system enables a dual chemo and photodynamic therapy to enhance the cancer therapy effect.


Asunto(s)
Doxorrubicina , Portadores de Fármacos , Ácido Hialurónico , Estructuras Metalorgánicas , Nanopartículas , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias/metabolismo , Neoplasias/patología
18.
Mol Pharm ; 16(6): 2522-2531, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31009223

RESUMEN

A multiarm nanomedicine template has been designed following bottom-up approach, which target neuropilin-1 (Nrp-1) receptor of cancer cells. Through this venture, we discovered that cucurbit [6] uril (CB [6]) binds with tubulin close to binding pocket of vinblastine site and perturbs tubulin polymerization. To increase the specificity of gold nanoparticle (GNP) toward Nrp-1-rich cancer cells, we further modified this GNP with Nrp-1 receptor-specific short peptide (CGNKRTR). Remarkably, we found an interesting self-assembly process upon addition of curcumin into the CB [6] and peptide-functionalized GNP, leading to the formation of a spherical nanocapsule (CGNP·Cur). It can deliver and release significantly higher amounts of anticancer drug curcumin in Nrp-1-rich cancer cells. It causes microtubule depolymerization and significant tumor regression in Nrp-1 overexpressed mice melanoma model. These interesting findings show that nanocapsule has high potential to develop a powerful anticancer nanomedicine and help in its preclinical validation.


Asunto(s)
Nanopartículas del Metal/química , Microtúbulos/metabolismo , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Nanomedicina/métodos , Neuropilina-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Microscopía por Crioelectrón , Oro/química , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Tubulina (Proteína)/metabolismo
19.
ACS Chem Neurosci ; 10(5): 2609-2620, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30840820

RESUMEN

Amyloid-beta (Aß) peptide misfolds into fibrillary aggregates (ß-sheet) and is deposited as amyloid plaques in the cellular environment, which severely damages intraneuronal connections leading to Alzheimer's disease (AD) pathogenesis. Furthermore, neurons are rich in tubulin/microtubules, and the intracellular network of microtubules also gets disrupted by the accumulation of Aß fiber in the brain. Hence, development of new potent molecules, which can simultaneously inhibit Aß fibrillations and stabilize microtubules, is particularly needed for the efficient therapeutic application in AD. To address these issues, here we introduced an innovative fusion strategy to design and develop next generation anti-AD therapeutic leads. This unexplored fusion strategy entails design and development of a potent nonapeptide by taking into account both the hydrophobic core (17-21) of Aß peptide and the taxol binding region of ß-tubulin. In vitro results suggest that this newly designed peptide interacts at the taxol binding region of ß-tubulin with a moderate binding affinity and promotes microtubule polymerization. It has the ability to bind at the hydrophobic core (17-21) of Aß, responsible for its aggregation, and prevent amyloid fibril as well as plaque formation. In addition, it interacts at the CAS site (catalytic anionic site) of acetylcholinesterase (AChE) and significantly inhibits AChE induced Aß fibrillation, stimulates neurite branching, and provides stability to intracellular microtubules and extensive protection of neurons against nerve growth factor (NGF) deprived neuron toxicity. Moreover, this newly designed peptide shows good stability in serum obtained from humans and efficiently permeates the blood-brain barrier (BBB) without showing any toxicity toward differentiated PC12 neurons as well as primary rat cortical neurons. This excellent feature of protecting the neurons by stabilizing the microtubules without showing any toxicity toward neurons will make this peptide a potent therapeutic agent of AD in the near future.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Tubulina (Proteína)/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Microtúbulos/efectos de los fármacos , Estructura Molecular , Fragmentos de Péptidos/metabolismo , Placa Amiloide/tratamiento farmacológico , Ratas , Tubulina (Proteína)/química
20.
Chem Commun (Camb) ; 55(16): 2356-2359, 2019 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-30724319

RESUMEN

An indole-rich tripodal microtubule inhibitor is designed, which binds at the DCVJ site of tubulin and inhibits its polymerization. It causes apoptotic death of cancer cells without affecting normal cells and inhibits the growth of tumors. Finally, STD-NMR and TR-NOESY experiments reveal that the indole appendages play a crucial role in interacting with tubulin.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...