Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Nanobiotechnology ; 22(1): 431, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39034407

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease with multifactorial etiology and intricate pathogenesis. In RA, repeated monotherapy is frequently associated with inadequate efficacy, drug resistance, and severe side effects. Therefore, a shift has occurred in clinical practice toward combination therapy. However, conventional combination therapy encounters several hindrances, including low selectivity to arthritic joints, short half-lives, and varying pharmacokinetics among coupled drugs. Emerging nanotechnology offers an incomparable opportunity for developing advanced combination therapy against RA. First, it allows for co-delivering multiple drugs with augmented physicochemical properties, targeted delivery capabilities, and controlled release profiles. Second, it enables therapeutic nanomaterials development, thereby expanding combination regimens to include multifunctional nanomedicines. Lastly, it facilitates the construction of all-in-one nanoplatforms assembled with multiple modalities, such as phototherapy, sonodynamic therapy, and imaging. Thus, nanotechnology offers a promising solution to the current bottleneck in both RA treatment and diagnosis. This review summarizes the rationale, advantages, and recent advances in nano-empowered combination therapy for RA. It also discusses safety considerations, drug-drug interactions, and the potential for clinical translation. Additionally, it provides design tips and an outlook on future developments in nano-empowered combination therapy. The objective of this review is to achieve a comprehensive understanding of the mechanisms underlying combination therapy for RA and unlock the maximum potential of nanotechnology, thereby facilitating the smooth transition of research findings from the laboratory to clinical practice.


Asunto(s)
Artritis Reumatoide , Humanos , Artritis Reumatoide/tratamiento farmacológico , Animales , Nanomedicina/métodos , Nanotecnología/métodos , Terapia Combinada , Antirreumáticos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Nanopartículas/química
2.
Mater Today (Kidlington) ; 38: 10-23, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33716549

RESUMEN

Systemic delivery of hydrophobic anti-cancer drugs with nanocarriers, particularly for drug-resistant and metastatic cancer, remain a challenge because of the difficulty to achieve high drug loading, while maintaining a small hydrodynamic size and colloid stability in blood to ensure delivery of an efficacious amount of drug to tumor cells. Here we introduce a new approach to address this challenge. In this approach, nanofibers of larger size with good drug loading capacity are first constructed by a self-assembly process, and upon intravascular injection and interacting with serum proteins in vivo, these nanofibers break down into ultra-fine nanoparticles of smaller size that inherit the drug loading property from their parent nanofibers. We demonstrate the efficacy of this approach with a clinically available anti-cancer drug: paclitaxel (PTX). In vitro, the PTX-loaded nanoparticles enter cancer cells and induce cellular apoptosis. In vivo, they demonstrate prolonged circulation in blood, induce no systemic toxicity, and show high potency in inhibiting tumor growth and metastasis in both mouse models of aggressive, drug-resistant breast cancer and melanoma. This study points to a new strategy toward improved anti-cancer drug delivery and therapy.

3.
Pharm Res ; 37(10): 197, 2020 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-32968837

RESUMEN

PURPOSE: To develop drug-combination nanoparticles (DcNPs) composed of hydrophilic gemcitabine (G) and hydrophobic paclitaxel (T) and deliver both drugs to metastatic cancer cells. METHODS: GT DcNPs were evaluated based on particle size and drug association efficiency (AE%). The effect of DcNP on GT plasma time-course and tissue distribution was characterized in mice and a pharmacokinetic model was developed. A GT distribution study into cancer nodules (derived from 4 T1 cells) was performed. RESULTS: An optimized GT DcNP composition (d = 59.2 nm ±9.2 nm) was found to be suitable for IV formulation. Plasma exposure of G and T were enhanced 61-fold and 3.8-fold when given in DcNP form compared to the conventional formulation, respectively. Mechanism based pharmacokinetic modeling and simulation show that both G and T remain highly associated to DcNPs in vivo (G: 98%, T:75%). GT DcNPs have minimal distribution to healthy organs with selective distribution and retention in tumor burdened tissue. Tumor bearing lungs had a 5-fold higher tissue-to-plasma ratio of gemcitabine in GT DcNPs compared to healthy lungs. CONCLUSIONS: DcNPs can deliver hydrophilic G and hydrophobic T together to cancer nodules and produce long acting exposure, likely due to stable GT association to DcNPs in vivo.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Combinación de Medicamentos , Nanopartículas/administración & dosificación , Metástasis de la Neoplasia/tratamiento farmacológico , Paclitaxel/farmacocinética , Animales , Desoxicitidina/sangre , Desoxicitidina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Femenino , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Paclitaxel/sangre , Bazo/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Gemcitabina
4.
Small ; 12(46): 6388-6397, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27671114

RESUMEN

Applications of hydrophobic drug-based nanocarriers (NCs) remain largely limited because of their low loading capacity. Here, development of a multifunctional hybrid NC made of a magnetic Fe3O4 core and a mesoporous silica shell embedded with carbon dots (CDs) and paclitaxel (PTX), and covered by another layer of silica is reported. The NC is prepared via a one-pot process under mild condition. The PTX loading method introduced in this study simplifies drug loading process and demonstrates a high loading capacity due to mesoporous silica dual-shell structure, supramolecular π-stacking between conjugated rings of PTX molecules, and aromatic rings of the CDs in the hybrid NC. The CDs serve as both confocal and two-photon fluorescence imaging probes, while the Fe3O4 core serves as a magnetic resonance imaging contrast agent. Significantly, NC releases PTX in response to near infrared irradiation as a result of local heating of the embedded CDs and the heating of CDs also provides an additional therapeutic effect by thermally killing cancer cells in tumor in addition to the chemotherapeutic effect of released PTX. Both in vitro and in vivo results show that NC demonstrates high therapeutic efficacy through a synergistic effect from the combined chemo-photothermal treatments.


Asunto(s)
Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Imagen Multimodal/métodos , Animales , Antineoplásicos/química , Línea Celular Tumoral , Doxorrubicina/química , Liberación de Fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones
5.
Bioconjug Chem ; 25(6): 1103-11, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24807049

RESUMEN

Molecularly targeted research and diagnostic tools are essential to advancing understanding and detection of many diseases. Metals often impart the desired functionality to these tools, and conjugation of high-affinity chelators to proteins is carried out to enable targeted delivery of the metal. This approach has been much more effective with large lanthanide series metals than smaller transition metals. Because chemical conjugation requires additional processing and purification steps and yields a heterogeneous mixture of products, inline incorporation of a peptide tag capable of metal binding is a highly preferable alternative. Development of a transition metal binding tag would provide opportunity to greatly expand metal-based analyses. The metal abstraction peptide (MAP) sequence was genetically engineered into recombinant protein to generate the claMP Tag. The effects of this tag on recombinant epidermal growth factor (EGF) protein expression, disulfide bond formation, tertiary structural integrity, and transition metal incorporation using nickel were examined to confirm the viability of utilizing the MAP sequence to generate linker-less metal conjugates.


Asunto(s)
Familia de Proteínas EGF/química , Níquel/química , Péptidos/química , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Familia de Proteínas EGF/farmacología , Humanos , Modelos Moleculares , Estructura Molecular , Níquel/farmacología , Péptidos/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas
6.
Environ Sci Technol ; 48(3): 1984-92, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24392862

RESUMEN

Some nanomaterials, such as Mg(OH)2 nanoflakes, are heavily used in pollutant adsorption and removal. Residues from these environmental remediations are potential hazardous materials. Safety evaluations of these materials are needed for environmental protection and human health. Although nanotoxicity has been widely investigated in recent years, research on the toxicity of nanoparticle/pollutant adducts has been rather inadequate. Here, we report the cellular perturbations and cytotoxicity of nano-Mg(OH)2/Cr(VI) adducts as a case study to elucidate how nanoparticle/pollutant adducts impact human cells. We found that Mg(OH)2 nanoflakes barely enter cells, while desorbed Cr(VI) anions enter cells, generate ROS, induce cell apoptosis, and cause cytotoxicity. This cytotoxicity is only a fraction of the cytotoxicity of free Cr(VI) because nano-Mg(OH)2 particles are able to retain more than half of their Cr(VI) anions.


Asunto(s)
Cromo/toxicidad , Hidróxido de Magnesio/toxicidad , Adsorción , Apoptosis/efectos de los fármacos , Cromo/metabolismo , Restauración y Remediación Ambiental , Células HEK293 , Células Hep G2 , Humanos , Hidróxido de Magnesio/metabolismo , Nanoestructuras , Estrés Oxidativo/efectos de los fármacos
7.
Front Oncol ; 14: 1393492, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756653

RESUMEN

Breast cancer with triple-negative subtype (TNBC) presents significant challenges with limited treatment options and a poorer prognosis than others. While PD-1/PD-L1 checkpoint inhibitors have shown promise, their efficacy in TNBC remains constrained. In recent years, nanoparticle (NP) technologies offer a novel approach to enhance cancer therapy by optimizing the tumor microenvironment and augmenting chemo- and immunotherapy effects in various preclinical and clinical settings. This review discusses recent investigations in NP strategies for improving PD-1/PD-L1 blockade-based combination therapy for TNBC. Those include single or multi-therapeutic NPs designed to enhance immunogenicity of the tumor, induce immunogenic cell death, and target immunosuppressive elements within the tumor microenvironment. The investigations also include NPs co-loaded with PD-L1 inhibitors and other therapeutic agents, leveraging targeted delivery and synergistic effects to maximize efficacy while minimizing systemic toxicity. Overall, NP approaches represent a promising avenue for enhancing PD-1/PD-L1 checkpoint blockade-based combination therapy in TNBC and encourage further developmental studies.

8.
PLoS One ; 19(3): e0300723, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38547077

RESUMEN

Preclinical models of cancer are vital for assessing and predicting efficacies and toxicities of novel treatments prior to testing in human subjects. Current pancreatic tumor models exhibit variable growth rates, unpredictable tumor size after implantation in non-native tissues, or require surgical implantation. Surgical implantation in the pancreas may produce not only unpredictable tumor uptake but could also elicit additional inflammatory responses. In searching for a pancreatic carcinoma cell that can be introduced into a mouse via simple injection, we found that Pan02, a murine ductal pancreatic adenocarcinoma derived from a pancreatic lesion of a C57BL/6 mouse, inoculated peritoneally can consistently produce pancreatic tumors. This intraperitoneal, but not intravenous, introduction of Pan02 cells leads to the attachment and growth of Pan02 in the pancreas before spreading to other tissues. Time-course tissue analysis indicates that the Pan02 cells first find, infiltrate, and grow within the pancreas, producing a pancreatic tumor model. This model appears to mimic pancreatic cancer development in humans and is the first reported use of Pan02 cells to produce orthotopic pancreatic and metastatic neoplasms in a mouse model without the need for tumor implantation within matrices or survival surgeries. This orthotopic pancreatic tumor model, with consistent tumor uptake, synchronized tumor development and survival, and predictable outcomes may enable and accelerate the preclinical evaluation of treatment candidates for pancreatic cancer.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Neoplasias Pancreáticas/patología , Adenocarcinoma/patología , Ratones Endogámicos C57BL , Carcinoma Ductal Pancreático/patología , Páncreas/patología , Modelos Animales de Enfermedad , Línea Celular Tumoral
9.
Pharmaceutics ; 15(3)2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36986876

RESUMEN

Leukemia remains incurable partly due to difficulties in reaching and maintaining therapeutic drug concentrations in the target tissues and cells. Next-generation drugs targeted to multiple cell checkpoints, including the orally active venetoclax (Bcl-2 target) and zanubrutinib (BTK target), are effective and have improved safety and tolerability compared to conventional, nontargeted chemotherapies. However, dosing with a single agent frequently leads to drug resistance; asynchronous coverage due to the peak-and-trough time-course of two or more oral drugs has prevented drug combinations from simultaneously knocking out the respective drugs' targets for sustained leukemia suppression. Higher doses of the drugs may potentially overcome asynchronous drug exposure in leukemic cells by saturating target occupancy, but higher doses often cause dose-limiting toxicities. To synchronize multiple drug target knockout, we have developed and characterized a drug combination nanoparticle (DcNP), which enables the transformation of two short-acting, orally active leukemic drugs, venetoclax and zanubrutinib, into long-acting nanoformulations (VZ-DCNPs). VZ-DCNPs exhibit synchronized and enhanced cell uptake and plasma exposure of both venetoclax and zanubrutinib. Both drugs are stabilized by lipid excipients to produce the VZ-DcNP nanoparticulate (d ~ 40 nm) product in suspension. The VZ-DcNP formulation has enhanced uptake of the two drugs (VZ) in immortalized leukemic cells (HL-60), threefold over that of its free drug counterpart. Additionally, drug-target selectivity of VZ was noted with MOLT-4 and K562 cells that overexpress each target. When given subcutaneously to mice, the half-lives of venetoclax and zanubrutinib were extended by approximately 43- and 5-fold, respectively, compared to an equivalent free VZ. Collectively, these data suggest that VZ in VZ-DcNP warrant consideration for preclinical and clinical development as a synchronized and long-acting drug-combination for the treatment of leukemia.

10.
Micromachines (Basel) ; 13(7)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35888961

RESUMEN

The assessment of the biodegradability of nanomaterials is of pragmatic importance for understanding the interactions between nanomaterials and biological systems and for the determination of ultimate fate of these materials as well as their potential use. We recently developed carbon-based biconcave nanodisks (CBBNs) serving as a versatile nanocarrier for enhanced accumulation in tumors and combined photothermal-chemotherapy. Here, we investigate both the enzymatic and cellular degradation of CBBNs by monitoring their cellular response with electron microscopy, near-infrared absorbance spectroscopy, and cell viability and oxidative stress assessments. Our results show that CBBNs underwent significant degradation in solutions catalyzed by horseradish peroxidase (HRP) and hydrogen peroxide (H2O2), or in the presence of macrophage cells. The ability of CBBNs to be degraded in biological systems provides suitability for their future biomedical applications.

11.
Pharmacol Ther ; 236: 108108, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34999182

RESUMEN

Despite advances in breast cancer treatments and related 5-year survival outcomes, metastatic breast cancer cures remain elusive. The current standard of care includes a combination of surgery, radiation therapy and drug therapy. However, even the most advanced procedures and treatments do not prevent breast cancer recurrence and metastasis. Once metastasis occurs, patient prognosis is poor. Recent elucidation of the spatiotemporal transit of metastatic cancer cells from primary tumor sites to distant sites provide an opportunity to integrate knowledge of drug disposition in our effort to enhance drug localization and exposure in cancer laden tissues . Novel technologies have been developed, but could be further refined to facilitate the distribution of drugs to target cancer cells and tissues. The purpose of this review is to highlight the challenges in metastatic breast cancer treatment and focus on novel drug combination and nanotechnology approaches to overcome the challenges. With improved definition of metastatic tissue target, directed localization and retention of multiple, pharmacologically active drugs to tissues and cells of interest may overcome the limitations in breast cancer treatment that may lead to a cure for breast cancer.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Nanopartículas/uso terapéutico , Nanotecnología/métodos , Recurrencia Local de Neoplasia
12.
J Am Chem Soc ; 133(35): 13918-21, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21827210

RESUMEN

To conquer complex and devastating diseases such as cancer, more coordinated and combined attack strategies are needed. We suggest that these can be beautifully achieved by using nanoconstruct design. We present an example showing that neuroblastoma cells are selectively killed by a nanoconstruct that specifically targets neuroblastoma cells, pushes cells to the vulnerable phase of the cell cycle, and greatly enhances radiation-induced cell death. The success of this multipronged attack approach launched by cell-embedded nanoconstructs demonstrates the power and flexibility of nanotechnology in treating cancer, a difficult task for a small molecule.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Neuroblastoma/terapia , Paclitaxel/administración & dosificación , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Nanomedicina/métodos , Nanoestructuras/análisis , Rayos X
14.
Front Bioeng Biotechnol ; 9: 646708, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33869157

RESUMEN

Understanding the basic interactions between engineered nanoparticles (ENPs) and biological systems is essential for evaluating ENPs' safety and developing better nanomedicine. Profound interactions between ENPs and biomolecules such as proteins are inevitable to occur when ENPs are administered or exposed to biological systems, for example, through intravenous injection, oral, or respiration. As a key component of these interactions, protein corona (PC) is immediately formed surrounding the outlayer of ENPs. PC formation is crucial because it gives ENPs a new biological identity by altering not only the physiochemical properties, but also the biobehaviors of ENPs. In the past two decades, most investigations about PC formation were carried out with in vitro systems which could not represent the true events occurring within in vivo systems. Most recently, studies of in vivo PC formation were reported, and it was found that the protein compositions and structures were very different from those formed in vitro. Herein, we provide an in-time review of the recent investigations of this in vivo PC formation of ENPs. In this review, commonly used characterization methods and compositions of in vivo PC are summarized firstly. Next, we highlight the impacts of the in vivo PC formation on absorption, blood circulation, biodistribution, metabolism, and toxicity of administered ENPs. We also introduce the applications of modulating in vivo PC formation in nanomedicine. We further discuss the challenges and future perspectives.

15.
Pharmaceutics ; 14(1)2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35056985

RESUMEN

Despite the availability of molecularly targeted treatments such as antibodies and small molecules for human epidermal growth factor receptor 2 (HER2), hormone receptor (HR), and programmed death-ligand 1 (PD-L1), limited treatment options are available for advanced metastatic breast cancer (MBC), which constitutes ~90% mortality. Many of these monotherapies often lead to drug resistance. Novel MBC-targeted drug-combination therapeutic approaches that may reduce resistance are urgently needed. We investigated intercellular adhesion molecule-1 (ICAM-1), which is abundant in MBC, as a potential target to co-localize two current drug combinations, gemcitabine (G) and paclitaxel (T), assembled in a novel drug-combination nanoparticle (GT DcNP) form. With an ICAM-1-binding peptide (referred to as LFA1-P) coated on GT DcNPs, we evaluated the role of the LFA1-P density in breast cancer cell localization in vitro and in vivo. We found that 1-2% LFA1-P peptide incorporated on GT DcNPs provided optimal cancer cell binding in vitro with ~4× enhancement compared to non-peptide GT DcNPs. The in vivo probing of GT DcNPs labeled with a near-infrared marker, indocyanine green, in mice by bio-imaging and G and T analyses indicated LFA1-P enhanced drug and GT DcNP localization in breast cancer cells. The target/healthy tissue (lung/gastrointestinal (GI)) ratio of particles increased by ~60× compared to the non-ligand control. Collectively, these data indicated that LFA1 on GT DcNPs may provide ICAM-1-targeted G and T drug combination delivery to advancing MBC cells found in lung tissues. As ICAM-1 is generally expressed even in breast cancers that are triple-negative phenotypes, which are unresponsive to inhibitors of nuclear receptors or HER2/estrogen receptor (ER) agents, ICAM-1-targeted LFA1-P-coated GT DcNPs should be considered for clinical development to improve therapeutic outcomes of MBCs.

16.
Biomater Sci ; 9(2): 471-481, 2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-32662460

RESUMEN

The ability to visualize and quantify apoptosis in vivo is critical to monitoring the disease response to treatment and providing prognostic information. However, the application of current apoptosis labeling probes faces significant challenges including nonspecific tissue uptake, inefficient apoptotic cell labeling and short monitoring windows. Here we report a highly specific apoptosis labeling nanoparticle (NP) probe with Pisum sativum agglutinin (PSA) as a tumor targeting ligand for prolonged in vivo apoptosis imaging. The NP (namely, IONP-Neu-PSA) consists of a magnetic iron oxide core (IONP) conjugated with PSA, and a reporter fluorophore. IONP-Neu-PSA demonstrated minimal cytotoxicity and high labeling specificity towards apoptotic cells in vitro. When applied in vivo, IONP-Neu-PSA tracks apoptotic tumors for a prolonged period of two weeks under near-IR imaging with low background noise. Moreover, IONP-Neu-PSA possesses T2 contrast enhancing properties that can potentially enable apoptosis detection by magnetic resonance imaging (MRI). The high specificity for apoptotic cells, sustained fluorescence signals, and non-invasive imaging capability exhibited by IONP-Neu-PSA make it a versatile tool for cancer treatment monitoring and pathological research.


Asunto(s)
Nanopartículas , Preparaciones Farmacéuticas , Apoptosis , Compuestos Férricos , Imagen por Resonancia Magnética
17.
Analyst ; 135(7): 1519-30, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20502814

RESUMEN

A significant increase in biomedical applications of nanomaterials and their potential toxicity demands versatile analytical techniques to determine protein-nanoparticle (NP) interactions. These diverse analytical techniques are reviewed. Spectroscopic methods play a significant role in studying binding affinity, binding ratio, and binding mechanisms. To elucidate NP-proteome interactions, chromatography and electrophoresis techniques are applied to separate NP-bound proteins and matrix assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) to identify these proteins. Since NP-protein binding is a dynamic event, surface plasmon resonance (SPR) and quartz crystal microbalance (QCM) are methods of choice to study the kinetics of NP-protein binding.


Asunto(s)
Nanopartículas/química , Proteínas/química , Electroforesis/métodos , Cinética , Espectrometría de Masas/métodos , Unión Proteica , Espectrofotometría/métodos , Resonancia por Plasmón de Superficie/métodos
18.
Nano Lett ; 9(12): 4370-5, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19902917

RESUMEN

We report our findings on cellular membrane penetration, endocytosis, endosomal leakage, and nuclear translocation of multiwalled carbon nanotubes (MWCNTs). Our data is consistent with a working model for MWCNTs' cell uptake and cellular translocations.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Endocitosis/fisiología , Endosomas/fisiología , Riñón/fisiología , Modelos Biológicos , Nanotubos de Carbono , Línea Celular , Humanos
19.
Nano Lett ; 9(6): 2280-4, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19408924

RESUMEN

Although carbon nanotubes (CNTs) have been shown to nonspecifically bind proteins through charge complementary, pi-pi stacking or hydrophobic interactions, they have not been shown to bind to a specific site on proteins. By generating surface molecular diversity, we created functionalized CNTs that recognize and bind to the catalytic site of alpha-chymotrypsin and inhibit its enzymatic activity competitively.


Asunto(s)
Quimotripsina/química , Nanotubos de Carbono/química , Dominio Catalítico , Técnicas Químicas Combinatorias , Sistemas de Liberación de Medicamentos , Fluorescencia , Interacciones Hidrofóbicas e Hidrofílicas
20.
Nanoscale Horiz ; 5(3): 573-579, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32118222

RESUMEN

Gadolinium (Gd)-based chelates are used as clinical T1 contrast agents for magnetic resonance imaging (MRI) due to their demonstrated high sensitivity and positive contrast enhancement capability. However, there has been an increasing safety concern about their use in medicine because of the toxicity of the metal ions released from these contrast agents when used in vivo. Although significant effort has been made in developing metal-free MRI contrast agents, none have matched the magnetic properties achieved by the gold standard clinical contrast agent, Gd diethylene penta-acetic acid (Gd-DTPA). Here, we report the development of a single-layer, boron-doped graphene quantum dot (termed SL-BGQD) that demonstrates better T1 contrast enhancement than Gd-DTPA. The SL-BGQD is shown to provide significantly higher positive contrast enhancement than the Gd-DTPA contrast agent in imaging vital organs, including kidneys, liver, and spleen, and especially, vasculatures. Further, our results show that the SL-BQGD is able to bypass the blood-brain barrier and allows sustained imaging for at least one hour with a single injection. Hematological and histopathological analyses show that the SL-BGQD demonstrates a non-toxic profile in wild-type mice and may, therefore, serve as an improved, safer alternative to currently available clinical MRI contrast agents.


Asunto(s)
Medios de Contraste/química , Imagen por Resonancia Magnética/métodos , Puntos Cuánticos/química , Animales , Barrera Hematoencefálica/metabolismo , Boro , Medios de Contraste/normas , Diagnóstico por Imagen/métodos , Diagnóstico por Imagen/normas , Grafito , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA