Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
Theranostics ; 14(6): 2605-2621, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646639

RESUMO

Rationale: Nab-paclitaxel (Abx) is widely employed in malignant tumor therapy. In tumor cells and pro-tumoral M2-type macrophages, the IL4 receptor (IL4R) is upregulated. This study aimed to elucidate the selective delivery of Abx to M2-type macrophages by targeting IL4R and reprogramming them into an anti-tumoral M1-type. Methods: Abx was conjugated with the IL4R-binding IL4RPep-1 peptide using click chemistry (IL4R-Abx). Cellular internalization, macrophage reprogramming and signal pathways, and tumor growth and metastasis by IL4R-Abx were examined. Results: IL4R-Abx was internalized into M2 macrophages more efficiently compared to the unmodified Abx and control peptide-conjugated Abx (Ctrl-Abx), which was primarily inhibited using an anti-IL4R antibody and a receptor-mediated endocytosis inhibitor compared with a macropinocytosis inhibitor. IL4R-Abx reprogrammed the M2-type macrophages into M1-like phenotype and increased reactive oxygen species (ROS) levels and extracellular release of high mobility group box 1 (HMGB1) in M2 macrophages at higher levels than Abx and Ctrl-Abx. The conditioned medium of IL4R-Abx-treated M2 macrophages skewed M2 macrophages into the M1-like phenotype, in which an anti-HMGB1 antibody and a toll-like receptor 4 (TLR4) inhibitor induced a blockade. IL4R-Abx accumulated at tumors, heightened immune-stimulatory cells while reducing immune-suppressing cells, and hampered tumor growth and metastasis in mice more efficiently than Abx and Ctrl-Abx. Conclusions: These results indicate that IL4R-targeting allows enhancement of M2-macrophage shaping into M1-like phenotype by Abx through the ROS-HMGB1-TLR4 axis, improvement of antitumor immunity, and thereby inhibition of tumor growth and metastasis, presenting a new approach to cancer immunotherapy.


Assuntos
Albuminas , Proteína HMGB1 , Macrófagos , Paclitaxel , Espécies Reativas de Oxigênio , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Proteína HMGB1/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Paclitaxel/farmacologia , Albuminas/metabolismo , Receptores de Interleucina-4/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/metabolismo , Camundongos Endogâmicos C57BL , Fenótipo , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Feminino
2.
Nat Commun ; 15(1): 1444, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365897

RESUMO

Transparent ultrasound transducers (TUTs) can seamlessly integrate optical and ultrasound components, but acoustic impedance mismatch prohibits existing TUTs from being practical substitutes for conventional opaque ultrasound transducers. Here, we propose a transparent adhesive based on a silicon dioxide-epoxy composite to fabricate matching and backing layers with acoustic impedances of 7.5 and 4-6 MRayl, respectively. By employing these layers, we develop an ultrasensitive, broadband TUT with 63% bandwidth at a single resonance frequency and high optical transparency ( > 80%), comparable to conventional opaque ultrasound transducers. Our TUT maximises both acoustic power and transfer efficiency with maximal spectrum flatness while minimising ringdowns. This enables high contrast and high-definition dual-modal ultrasound and photoacoustic imaging in live animals and humans. Both modalities reach an imaging depth of > 15 mm, with depth-to-resolution ratios exceeding 500 and 370, respectively. This development sets a new standard for TUTs, advancing the possibilities of sensor fusion.


Assuntos
Técnicas Fotoacústicas , Humanos , Técnicas Fotoacústicas/métodos , Transdutores , Desenho de Equipamento , Ultrassonografia , Impedância Elétrica
3.
Biomater Sci ; 12(6): 1477-1489, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38294258

RESUMO

Osteoarthritis (OA) is a degenerative arthritis disease marked by inflammation, pain, and cartilage deterioration. Elevated nitric oxide (NO) levels play a pivotal role in mediating OA-related inflammation and are found in abundance within OA joints. This study introduces a NO-scavenging hyaluronic acid conjugate (HA-NSc) bearing both lubrication and anti-inflammatory properties for the treatment of osteoarthritis. For this, a derivative of o-phenylenediamine (o-PD) with good NO-scavenging capability (NSc) is designed, synthesized and chemically conjugated to HA. Owing to the amphiphilicity, this as-synthesized HA-NSc conjugate formed self-assembled nanoparticles (HA-NSc NPs) under aqueous conditions. When treated with activated murine macrophage RAW 264.7 cells that produce high levels of NO, these nanoparticles effectively reduced intracellular NO concentrations and inflammatory cytokines. In an OA animal model, the HA-NSc NPs significantly alleviated pain and diminished the cartilage damage due to the combined lubricating property of HA and NO-scavenging ability of NSc. Overall, the results from the study suggest HA-NSc NPs as a dual-action therapeutic agent for the treatment of OA by alleviating pain, inflammation, and joint damage, and also positioning the HA-NSc NPs as a promising candidate for innovative treatment of OA.


Assuntos
Cartilagem Articular , Nanopartículas , Osteoartrite , Camundongos , Animais , Ácido Hialurônico/química , Óxido Nítrico , Inflamação/tratamento farmacológico , Dor/tratamento farmacológico , Osteoartrite/tratamento farmacológico
4.
Biomater Sci ; 12(4): 1031-1041, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38214329

RESUMO

This study presents a novel approach for the development of DNA-functionalized gold nanoparticles (AuNPs) capable of responding to disease-specific factors and microenvironmental changes, resulting in an effective anti-tumor effect via photothermal therapy. The AuNPs are decorated with two types of DNAs, an i-motif duplex and a VEGF split aptamer, enabling recognition of changes in pH and VEGF, respectively. The formation of VEGF aptamers on the AuNPs induces their aggregation, further enhanced by VEGF ligands. The resulting changes in the optical properties of the AuNPs are detected by monitoring the absorbance. Upon irradiation with a near-infrared laser, the aggregated AuNPs generate heat due to their thermoplasmonic characteristic, leading to an anti-tumor effect. This study demonstrates the enhanced anti-tumor effect of DNA-functionalized AuNPs via photothermal therapy in both in vitro and in vivo tumor models. These findings suggest the potential utilization of such functional AuNPs for precise disease diagnosis and treatment by detecting disease-related factors in the microenvironment.


Assuntos
Nanopartículas Metálicas , Neoplasias , Humanos , Fator A de Crescimento do Endotélio Vascular , Ouro/química , Nanopartículas Metálicas/química , Neoplasias/tratamento farmacológico , DNA , Concentração de Íons de Hidrogênio , Microambiente Tumoral
5.
Nanoscale ; 16(2): 742-751, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38086680

RESUMO

The approach of using ferroptosis to treat cancer has garnered attention due to its promising potential. However, the effectiveness of this therapy is often limited by the inherent redox system in cancer cells and the presence of ferritin as an iron ion storage molecule. To address this issue, we have designed a polymeric iron oxide nanocomplex loaded with sulfasalazine as a ferritinophagy-assisted ferroptosis inducing agent. The nanocomplex is based on a pH-responsive drug releasing platform that enables improved ferroptosis anti-cancer therapy. The nanocomplex was synthesized using polymerized phenylboronic acid decorated with iron oxide and further loaded with sulfasalazine by interacting with polymerized phenylboronic acid. Upon entering cancer cells, the nanocomplex releases sulfasalazine at the lysosomal acidic pH, which causes the complex to degrade into the labile iron ion (Fe2+). This process inhibits the production of GSH and reproduces the labile iron ion by degrading ferritin. As a result, an excess iron ion pool is formed, and the facilitated Fenton reaction induces an improved ferroptosis anti-cancer effect. Moreover, our research has demonstrated that the nanocomplex effectively regresses tumors, thereby representing significant inhibition of tumor growth using in vivo models. We believe that this ferritinophagy-assisted ferroptosis strategy using the nanocomplex provides a promising solution for iron-based anti-cancer therapy.


Assuntos
Ferroptose , Sulfassalazina , Sulfassalazina/farmacologia , Ferro , Ferritinas
6.
J Control Release ; 363: 496-506, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37788761

RESUMO

Indoleamine 2,3-dioxygenase (IDO) has been studied as a promising target for cancer immunotherapy. IDO catalyzes the oxidation of tryptophan into kynurenine, which subsequently activates regulatory T cells, thereby promoting an immunosuppressive microenvironment in the tumor tissue. Due to its overexpression in tumor cells, IDO itself could be a tumor-specific stimulus for targeted cancer therapy. Toward this objective, we developed IDO-triggered swellable micelles for targeted cancer immunotherapy. The micelles are prepared by the self-assembly of amphiphilic polymers containing polymerized tryptophan as a hydrophobic block. The micelles exhibited IDO-responsive behavior via solubility conversion of the hydrophobic core triggered by the oxidation of tryptophan residues into kynurenine. The micelles were internalized into tumor cells and disassembled by overexpressed IDO. Loaded with IDO inhibitor, the micelle presented enhanced therapeutic antitumor effect, and effector T-cells were recruited into the tumor tissue. We demonstrated that overexpressed IDO in cancer cells could be utilized as a tumor-specific stimulus, and utilizing an IDO-responsive drug delivery system holds great promise for targeted cancer therapy and immunomodulation.


Assuntos
Neoplasias , Triptofano , Humanos , Triptofano/química , Triptofano/farmacologia , Cinurenina , Micelas , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Imunoterapia , Microambiente Tumoral
7.
Small ; 19(50): e2303668, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37612796

RESUMO

Transcytosis is an active transcellular transportation pathway that has garnered interest for overcoming the limited deep penetration of nanomedicines in solid tumors. In this study, a charge-convertible nanomedicine that facilitates deep penetration into solid tumors via transcytosis is designed. It is an albumin-based calcium phosphate nanomedicine loaded with IR820 (mAlb-820@CaP) for high-resolution photoacoustic imaging and enhanced photothermal therapy. Biomineralization on the surface stabilizes the albumin-IR820 complex during circulation and provides calcium ions (Ca2+ ) for tissue penetration on degradation in an acidic environment. pH-triggered transcytosis of the nanomedicine enabled by caveolae-mediated endocytosis and calcium ion-induced exocytosis in 2D cellular, 3D spheroid, and in vivo tumor models is demonstrated. Notably, the extravasation and penetration ability of the nanomedicine is observed in vivo using a high-resolution photoacoustic system, and nanomedicine shows the most potent photothermal antitumor effect in vivo. Overall, the strategy provides a versatile theragnosis platform for both noninvasive photoacoustic imaging and high therapeutic efficiency resulting from deep penetration of nanomedicine.


Assuntos
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Humanos , Nanomedicina , Cálcio/metabolismo , Nanomedicina Teranóstica/métodos , Linhagem Celular Tumoral , Nanopartículas/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia/métodos , Transcitose , Albuminas/metabolismo , Técnicas Fotoacústicas/métodos
8.
Biofabrication ; 15(3)2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37336204

RESUMO

Fused deposition modeling (FDM) is a three-dimensional (3D) printing technology typically used in tissue engineering. However, 3D-printed row scaffolds manufactured using material extrusion techniques have low cell affinity on the surface and an insufficient biocompatible environment for desirable tissue regeneration. Thus, in this study, plasma treatment was used to render surface modification for enhancing the biocompatibility of 3D-printed scaffolds. We designed a plasma-based 3D printing system with dual heads comprising a plasma device and a regular 3D FDM printer head for a layer-by-layer nitrogen plasma treatment. Accordingly, the wettability, roughness, and protein adsorption capability of the 3D-printed scaffold significantly increased with the plasma treatment time. Hence, the layer-by-layer plasma-treated (LBLT) scaffold exhibited significantly enhanced cell adhesion and proliferation in anin vitroassay. Furthermore, the LBLT scaffold demonstrated a higher tissue infiltration and lower collagen encapsulation than those demonstrated by a non-plasma-treated scaffold in anin vivoassay. Our approach has great potential for various tissue-engineering applications via the adjustment of gas or precursor levels. In particular, this system can fabricate scaffolds capable of holding a biocompatible surface on an entire 3D-printed strut. Thus, our one-step 3D printing approach is a promising platform to overcome the limitations of current biocompatible 3D scaffold engineering.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Engenharia Tecidual/métodos , Colágeno , Adesão Celular , Impressão Tridimensional
9.
EMBO Rep ; 24(8): e56538, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37377038

RESUMO

The ER regulates the spatiotemporal organization of endolysosomal systems by membrane contact. In addition to tethering via heterotypic interactions on both organelles, we present a novel ER-endosome tethering mechanism mediated by homotypic interactions. The single-pass transmembrane protein SCOTIN is detected in the membrane of the ER and endosomes. In SCOTIN-knockout (KO) cells, the ER-late endosome contacts are reduced, and the perinuclear positioning of endosomes is disturbed. The cytosolic proline-rich domain (PRD) of SCOTIN forms homotypic assemblies in vitro and is necessary for ER-endosome membrane tethering in cells. A region of 28 amino acids spanning 150-177 within the SCOTIN PRD is essential to elicit membrane tethering and endosomal dynamics, as verified by reconstitution in SCOTIN-KO cells. The assembly of SCOTIN (PRD) is sufficient to mediate membrane tethering, as purified SCOTIN (PRD), but not SCOTIN (PRDΔ150-177), brings two different liposomes closer in vitro. Using organelle-specific targeting of a chimeric PRD domain shows that only the presence on both organellar membranes enables the ER-endosome membrane contact, indicating that the assembly of SCOTIN on heterologous membranes mediates organelle tethering.


Assuntos
Retículo Endoplasmático , Membranas Intracelulares , Membranas Intracelulares/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Endossomos/metabolismo
10.
J Control Release ; 360: 940-952, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37001565

RESUMO

Owing to a lack of reliable markers and therapeutic targets, pancreatic ductal adenocarcinoma (PDAC) remains the most lethal malignant tumor despite numerous therapeutic advances. In this study, we utilized cell-SELEX to isolate a DNA aptamer recognizing the natural conformation of the target on the cell surface. PAp7T8, an aptamer optimized by size and chemical modification, exhibited specific targeting to pancreatic cancer cells and orthotopic xenograft pancreatic tumors. To confer therapeutic functions to the aptamer, we adopted a drug-conjugated oligobody (DOligobody) strategy. Monomethyl auristatin E was used as a cytotoxic drug, digoxigenin acted as a hapten, and the humanized anti-digoxigenin antibody served as a universal carrier of the aptamer. The resulting PAp7T8-DOligobody showed extended in vivo half-life and markedly inhibited tumor growth in an orthotopic pancreatic cancer xenograft model without causing significant toxicity. Therefore, PAp7T8-DOligobody represents a promising novel therapeutic delivery platform for PDAC.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Preparações Farmacêuticas , Linhagem Celular Tumoral , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Anticorpos , Oligonucleotídeos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
11.
Biomater Sci ; 11(7): 2395-2404, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36786425

RESUMO

Nitric oxide (NO) plays various physiologically favorable roles in the body. However, excessive production of NO causes inflammation and leads to various chronic inflammatory diseases. A typical NO-related disease is rheumatoid arthritis (RA), and it is well known that NO is a critical molecule for inflammation in the pathophysiology of RA. Therefore, depletion of NO can be an attractive treatment option for RA. In this study, we proposed a new method to discover effective NO scavengers in the form of small molecules. o-Phenylenediamine (o-PD), the core structure of the NO scavenger, is a diamino-aromatic compound that irreversibly reacts with NO through nucleophilic substitution of amine. Inspired by the nucleophilicity, we attempted to find new scavenger candidates by searching for conditions that increase the nucleophilicity of the amine moieties. Candidates were classified into the basic form o-PD, monoamine aniline, o-PD substituted with a nitro group, carboxyl group, and three methyl groups. The NO-scavenging ability of these candidates was demonstrated using the DAF-2 assay. N-Methyl-o-PD (N-Me) in the methyl (-CH3) group had the highest reactivity with NO among the candidates, and the efficiency of NO scavengers was confirmed in vitro and in vivo. Depleted levels of NO and reduced levels of pro-inflammatory cytokines by N-Me demonstrated remarkable therapeutic efficacy against joint damage and delayed severity in a collagen-induced arthritis (CIA) model. Therefore, our findings suggest that N-Me is a new NO scavenger with great potential for RA treatment and further clinical drug development.


Assuntos
Artrite Reumatoide , Óxido Nítrico , Humanos , Artrite Reumatoide/tratamento farmacológico , Inflamação/tratamento farmacológico , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Citocinas
12.
Nat Biomed Eng ; 7(2): 149-163, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36456857

RESUMO

Deep brain stimulation via implanted electrodes can alleviate neuronal disorders. However, its applicability is constrained by side effects resulting from the insertion of electrodes into the brain. Here, we show that systemically administered piezoelectric nanoparticles producing nitric oxide and generating direct current under high-intensity focused ultrasound can be used to stimulate deep tissue in the brain. The release of nitric oxide temporarily disrupted tight junctions in the blood-brain barrier, allowing for the accumulation of the nanoparticles into brain parenchyma, and the piezoelectrically induced output current stimulated the release of dopamine by dopaminergic neuron-like cells. In a mouse model of Parkinson's disease, the ultrasound-responsive nanoparticles alleviated the symptoms of the disease without causing overt toxicity. The strategy may inspire the development of other minimally invasive therapies for neurodegenerative diseases.


Assuntos
Estimulação Encefálica Profunda , Nanopartículas , Camundongos , Animais , Barreira Hematoencefálica , Óxido Nítrico , Estimulação Encefálica Profunda/métodos , Encéfalo
13.
Adv Sci (Weinh) ; : e2202089, 2022 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36354200

RESUMO

Photoacoustic computed tomography (PACT) has become a premier preclinical and clinical imaging modality. Although PACT's image quality can be dramatically improved with a large number of ultrasound (US) transducer elements and associated multiplexed data acquisition systems, the associated high system cost and/or slow temporal resolution are significant problems. Here, a deep learning-based approach is demonstrated that qualitatively and quantitively diminishes the limited-view artifacts that reduce image quality and improves the slow temporal resolution. This deep learning-enhanced multiparametric dynamic volumetric PACT approach, called DL-PACT, requires only a clustered subset of many US transducer elements on the conventional multiparametric PACT. Using DL-PACT, high-quality static structural and dynamic contrast-enhanced whole-body images as well as dynamic functional brain images of live animals and humans are successfully acquired, all in a relatively fast and cost-effective manner. It is believed that the strategy can significantly advance the use of PACT technology for preclinical and clinical applications such as neurology, cardiology, pharmacology, endocrinology, and oncology.

14.
Nano Converg ; 9(1): 47, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36214916

RESUMO

Bone healing involves complex processes including inflammation, induction, and remodeling. In this context, anti-inflammatory and osteoconductive multi-functional nanoparticles have attracted considerable attention for application in improved bone tissue regeneration. In particular, nanoparticles that promote suppression of inflammatory response after injury and direction of desirable tissue regeneration events are of immense interest to researchers. We herein report a one-step method to prepare multi-functional nanoparticles using tannic acid (TA) and simulated body fluid (SBF) containing multiple mineral ions. Mineral-tannic acid nanoparticles (mTNs) were rapidly fabricated in 10 min, and their size (around 250-350 nm) and chemical composition were controlled through the TA concentration. In vitro analysis using human adipose derived stem cells (hADSCs) showed that mTNs effectively scavenged reactive oxygen species (ROS) and enhanced osteogenesis of hADSCs by inducing secretion of alkaline phosphatase. mTNs also increased osteogenic marker gene expression even in the presence of ROS, which can generally arrest osteogenesis (OPN: 1.74, RUNX2: 1.90, OCN: 1.47-fold changes relative to cells not treated with mTNs). In vivo analysis using a mouse peritonitis model revealed that mTNs showed anti-inflammatory effects by decreasing levels of pro-inflammatory cytokines in blood (IL-6: 73 ± 4, TNF-α: 42 ± 2%) and peritoneal fluid (IL-6: 78 ± 2, TNF-α: 21 ± 6%). We believe that this one-step method for fabrication of multi-functional nanoparticles has considerable potential in tissue engineering approaches that require control of complex microenvironments, as required for tissue regeneration.

15.
Des Monomers Polym ; 25(1): 245-253, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36017475

RESUMO

Carbon-based nanomaterials, such as carbon nanotubes, fullerenes, nanodiamonds, and graphene, have been investigated for various biomedical applications, including biological imaging, photothermal therapy, drug/gene delivery, cancer therapy, biosensors, and electrochemical sensors. Graphene oxide (GO) has unique physicochemical properties and can be used to restore conductivity through oxidation. In this study, we developed poly(N-isopropylacrylamide) (PNIPAM)-based nanogel systems containing GO for controlled in vitro drug delivery. The photothermal effects of the PNIPAM/GO- and PNIPAMAAM/GO-based nanogel systems were enhanced. The release of DOX from the PNIPAM/GO-based nanogel was achieved using the photothermal effect of near-infrared irradiation. Using a Cell Counting Kit-8 assay, the cytotoxicity of all conditions demonstrated that the PNIPAM composite-based nanogels were biocompatible with no significance.

16.
Biosens Bioelectron ; 216: 114612, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-35952434

RESUMO

Theranostic, which integrates the diagnosis and tumor treatment in tandem, is an emerging strategy in cancer treatment. Here, we report a novel and unique theranostic nanoparticle, HBCP NP, based on hexa-BODIPY cyclophosphazene (HBCP). Due to the unique bulky molecular structure of HBCP, this nanoparticle can simultaneously perform near-infrared (NIR) fluorescence imaging and photoacoustic imaging (PAI). Interestingly, since reactive oxygen species (ROS) generation of HBCP NPs is completely inhibited, 'safe' fluorescence imaging is possible without the risk of cell damage even under laser irradiation. Finally, NIR fluorescence imaging and PAI in 4T1 tumor-bearing mice demonstrated selective accumulation of HBCP NPs at tumor sites. In addition, HBCP NPs exhibited excellent photothermal effects under high-power laser irradiation, achieving effective tumor growth inhibition.


Assuntos
Técnicas Biossensoriais , Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Animais , Compostos de Boro , Linhagem Celular Tumoral , Hexosaminidase A , Camundongos , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Imagem Óptica , Técnicas Fotoacústicas/métodos , Espécies Reativas de Oxigênio , Nanomedicina Teranóstica/métodos
17.
Acc Chem Res ; 55(17): 2384-2396, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-35786846

RESUMO

Nitric oxide (NO) is an endogenous signaling molecule that participates in various physiological and biological pathways associated with vasodilation, immune response, and cell apoptosis. Interestingly, NO has versatile and distinct functions in vivo depending on its concentration and the duration of exposure; it aids cellular proliferation at nanomolar concentrations but causes cellular death at micromolar concentrations. Therefore, achieving the precise and on-demand modulation of microenvironmental NO concentrations has become a major research target in biomedical fields. To this end, many studies have investigated feasible means for developing functional moieties that can either exogenously donate or selectively scavenge NO. However, these advances are limited by poor stability and a lack of target specificity, which represent two significant obstacles regarding the spatiotemporal adjustment of NO in vivo. Our group has addressed this issue by contributing to the development of next-generation NO-modulatory materials over the past decade. Over this period, we utilized various polymeric, inorganic, and hybrid systems to enhance the bioavailability of traditional NO donors or scavengers in an attempt to maximize their clinical usage while also minimizing their unwanted side effects. In this Account, strategies regarding the rational design of NO-modulatory materials are first summarized and discussed, depending on their specific purposes. These strategies include chemical approaches for encapsulating traditional NO donors inside specific vehicles; this prevents spontaneous NO release and allows said donors to be exposed on-demand, under a certain stimulus. The current status of these approaches and the recent contributions of other groups are also comprehensively discussed here to ensure an objective understanding of the topic. Moreover, in this paper, we discuss strategies for the selective depletion of NO from local inflammatory sites, where the overproduction of NO is problematic. Finally, the major challenges for current NO-modulatory systems are discussed, and requirements are outlined that need to be tackled to achieve their future therapeutic development. Starting from this current, relatively early stage of development, we propose that, through continuous efforts to surmount existing challenges, it will be possible in the future to achieve clinical translations regarding NO-modulatory systems. This Account provides insightful guidelines regarding the rational design of NO-modulatory systems for various biomedical applications. Moreover, it can facilitate the achievement of previously unattainable goals while revolutionizing future therapeutics.


Assuntos
Óxido Nítrico , Polímeros , Óxido Nítrico/metabolismo
18.
Anticancer Res ; 42(7): 3463-3473, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35790297

RESUMO

BACKGROUND/AIM: Locoregional treatments for hepatocellular carcinoma (HCC) induce immunogenic cell death and a tumor-specific immune response, but infiltration and activation of immune cells in the liver have not been clearly described. Transarterial chemoembolization (TACE) or transarterial chemotherapy (TAC) without embolization have been used to treat intermediate or advanced stage HCC patients. The identification of intrahepatic immune cell changes after locoregional therapy provides a theoretical basis for the combination with immune checkpoint inhibitors (ICIs) in HCC. This study aimed to determine the anticancer effect and changes in the liver immune cell population and function after direct injection of polymerized phenylboronic acid-conjugated doxorubicin (pPBA-Dox) nanocomplexes into the liver through TAC. MATERIALS AND METHODS: pPBA-Dox nanocomplexes were delivered directly to the liver cancer in a rat model by transarterial methods. Anticancer effect was confirmed by magnetic resonance imaging (MRI), and the immune cell population and functional changes were confirmed by flow cytometry (FACS). RESULTS: We first established a rat liver cancer model by implanting McA-RH7777 into rats and confirmed the formation of liver cancer through MRI, pathological examinations, and biochemical tests. Transarterial injection of pPBA-Dox nanocomplexes had a stronger anticancer effect than conventional Dox alone. Higher numbers of CD8+ and CD4+ T cells with activated phenotypes were infiltrated into the tumor microenvironment after transarterial pPBA-Dox treatments than after Dox alone treatment, suggesting the induction of stronger local immune responses by pPBA-Dox than Dox alone. CONCLUSION: This study provides a theoretical basis for TAC combined with ICIs and insight into novel targeted therapies using nanocomplexes for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Quimioembolização Terapêutica , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Quimioembolização Terapêutica/métodos , Doxorrubicina , Imunidade , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Ratos , Microambiente Tumoral
19.
Macromol Biosci ; 22(10): e2200162, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35895972

RESUMO

Synthetic scaffolds, as bone grafts, provide a favorable environment for the repair and growth of new bone tissue at defect sites. However, the lack of angio- and osteo-induction limits the usefulness of artificial scaffolds for bone regeneration. Nitric oxide (NO) performs essential roles in healing processes, such as regulating inflammation and addressing incomplete revascularization. In this study, a polymer capable of controlled NO release is developed to promote the osteogenic capacity in artificial scaffolds. The biological efficiency of the NO compound is assessed by its effect on pre-osteoblasts and macrophages in vitro and the extent of vascularization and bone formation in the calvaria defect model in vivo. The compound does not inhibit cell adhesion or proliferation. NO treatment significantly increases both alkaline phosphatase activity and mineralization in pre-osteoblasts. Macrophages treated with NO secrete high levels of anti-inflammatory factors and adopt the pro-regenerative phenotype. In the critical-sized defect model, the collagen scaffold containing the NO compound enhances neovascularization and bone formation. The developed NO-releasing system promotes osteogenesis and regeneration of damaged bone tissue. As the multiple functions of NO involve macrophage modulation and angiogenesis, such release systems may be valuable for guiding bone regeneration in critical-sized defects.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Fosfatase Alcalina/metabolismo , Regeneração Óssea , Diferenciação Celular , Colágeno/metabolismo , Óxido Nítrico/metabolismo , Osteogênese/genética , Polímeros/metabolismo , Polímeros/farmacologia , Alicerces Teciduais
20.
Altern Ther Health Med ; 28(7): 95-103, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35687702

RESUMO

Context: Aging is connected to a decline in muscular strength, flexibility, and agility. Some studies have found that resistance exercise using an elastic band can prevent chronic health problems such as osteoporosis, degenerative diseases, and frozen shoulders. Objective: The study intended to investigate the effects of a 12-week program of resistance exercise using an elastic band on the pain, stress, range of motion (ROM), and body composition of older adults. Design: The research team designed a randomized controlled trial (RCT). Setting: The study took place in Gimcheon City in the Republic of Korea. Participants: Participants were 80 adults aged 60 or older residing in the community. Intervention: Participants were randomly assigned using random number generation to an intervention group (n = 40) or a control group (n = 40). The intervention group participated in a resistance exercise program using an elastic band three days a week for 12 weeks. The control group followed a daily routine for 12 weeks. Outcome Measures: Measurements were conducted three times: at baseline, at the sixth week of treatment, and postinterventon. Results: Regarding participants' general characteristics and homogeneity of the dependent variables at baseline, no significant differences existed between the groups. Postintervention, 6 weeks a significant difference in stress (F = -4.02, P < .001) between the two groups. Moreover, significant variances in the shoulders' ROM (left, F = 3.40, P < .001; and right, F = 3.83, P < .001) a significant difference. 12 weeks a significant difference in shoulder pain existed (F = 19.58, P < .001), and stress (F = 15.36, P < .001) between the two groups. Moreover, significant variances in the shoulders' ROM (left, F = 4.63, P < .001; and right, F = 5.30, P < .001), as well as in the thickness of muscles (left, F = 5.55, P < .001; and right, F = 3.10, P = .003), between the two groups. As a result of measuring the right fat thickness, a significant difference in the target area was also found (left, F = -2.748, P = .008; and right, F = -3.13, P = .002). Conclusions: The resistance exercise that participants performed gradually reduced participants' shoulder pain and stress, improved their shoulders' ROM, and increased muscle mass around the shoulder joint. Therefore, the program can be recommended for adults aged 60 or older complaining of shoulder pain, to reduce shoulder pain and stress, improve joint ROM, and enhance body composition around the shoulder joint.


Assuntos
Treinamento Resistido , Dor de Ombro , Idoso , Composição Corporal , Exercício Físico , Terapia por Exercício , Humanos , Amplitude de Movimento Articular/fisiologia , Dor de Ombro/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...